1
|
Lu G, Zhang L, Zhang Y, Wang J, Zhou X, Fang X, Ma Z. Preparation of accelerated-wound-healing lignin/dopamine-based nano-Fe 3O 4 hydrogels in sensing. Int J Biol Macromol 2024; 280:135942. [PMID: 39322138 DOI: 10.1016/j.ijbiomac.2024.135942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/26/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Flexible conductive hydrogels hold great promise for applications in motion and medical detection. It is difficult to produce conductive hydrogel epidermal sensors in wearable hydrogels with dependable adhesion, sensing, and wound-healing properties. Nano-Fe3O4 was used as physical cross-linking points in the polyacrylamide/polyvinyl alcohol double network (PP) to increase the strain capacity of the hydrogel. The conductive lignin-dopamine (LD) was immobilized on the surface of Fe3O4 particles, and the LD-coated Fe3O4 was then incorporated into the double network hydrogel to create the PP/LD/Fe3O4 hydrogel. This work was done to look into the possibility of using Fe3O4 hydrogels as flexible strain sensors. The addition of LD/Fe3O4 caused the composite hydrogel to strain up to 124 %, with a modulus of elasticity of 21,308 Pa and electrical conductivity as high as 2.3 S•m-1 following the introduction of LD/Fe3O4. Moreover, the PP/LD/Fe3O4 hydrogel's adhesive qualities offered adequate antimicrobial properties and promoted wound healing. These results indicate that the developed electricity-responsive and tissue-adhesive hydrogel dressing offers a candidate to serve as a tissue sealant for wound healing.
Collapse
Affiliation(s)
- Geng Lu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of Emergency Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lisha Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yue Zhang
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Jun Wang
- Department of Emergency Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xin Zhou
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xiang Fang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Department of Emergency Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Li Z, Qin B, Liu H, Du S, Liu Y, He L, Xu B, Du L. Mesoporous silica thin film as effective coating for enhancing osteogenesis through selective protein adsorption and blood clotting. Biomed Mater 2024; 19:055040. [PMID: 39094621 DOI: 10.1088/1748-605x/ad6ac2] [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: 05/31/2024] [Accepted: 08/02/2024] [Indexed: 08/04/2024]
Abstract
The role of blood clots in tissue repair has been identified for a long time; however, its participation in the integration between implants and host tissues has attracted attention only in recent years. In this work, a mesoporous silica thin film (MSTF) with either vertical or parallel orientation was deposited on titania nanotubes surface, resulting in superhydrophilic nanoporous surfaces. A proteomic analysis of blood plasma adsorption revealed that the MSTF coating could significantly increase the abundance of acidic proteins and the adsorption of coagulation factors (XII and XI), with the help of cations (Na+, Ca2+) binding. As a result, both the activation of platelets and the formation of blood clots were significantly enhanced on the MSTF surface with more condensed fibrin networks. The two classical growth factors of platelets-derived growth factors-AB and transformed growth factors-βwere enriched in blood clots from the MSTF surface, which accounted for robust osteogenesis bothin vitroandin vivo. This study demonstrates that MSTF may be a promising coating to enhance osteogenesis by modulating blood clot formation.
Collapse
Affiliation(s)
- Zhe Li
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
- Department of Digital Oral Implantology and Prothodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| | - Bowen Qin
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Huan Liu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
- Department of Implant Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| | - Shimin Du
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
- Department of Implant Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| | - Yunxian Liu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
- Department of Implant Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| | - Lixing He
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
- Department of Digital Oral Implantology and Prothodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| | - Boya Xu
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
- Department of Digital Oral Implantology and Prothodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| | - Liangzhi Du
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
- Department of Digital Oral Implantology and Prothodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, People's Republic of China
| |
Collapse
|
3
|
Xiao M, Wang L, Tang Q, Yang Q, Yang X, Zhu G, Lei L, Li S. Postoperative tumor treatment strategies: From basic research to clinical therapy. VIEW 2024; 5. [DOI: 10.1002/viw.20230117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/15/2024] [Indexed: 07/04/2024] Open
Abstract
AbstractDespite progression in advanced treatments for malignant tumors, surgery remains the primary treatment intervention, which removes a large portion of firm tumor tissues; however, the postoperative phase poses a possible risk for provincial tumor recurrence and metastasis. Consequently, the prevention of tumor recurrence and metastasis has attracted research attention. In this review, we summarized the postoperative treatment strategies for various tumors from both basic research and clinical perspectives. We delineated the underlying factors contributing to the recurrence of malignant tumors with a substantial prevalence rate, related molecular mechanisms of tumor recurrence post‐surgery, and related means of monitoring recurrence and metastasis after surgery. Furthermore, we described relevant therapeutic approaches for postoperative tumor recurrence, including chemotherapy, radiation therapy, immunotherapy, targeted therapy, and photodynamic therapy. This review focused on the emerging technologies used for postoperative tumor treatment in recent years in terms of functional classification, including the prevention of postoperative tumor recurrence, functional reconstruction, and monitoring of recurrence. Finally, we discussed the future development and deficiencies of postoperative tumor therapy. To understand postoperative treatment strategies for tumors from clinical treatment and basic research and further guide the research directions for postoperative tumors.
Collapse
Affiliation(s)
- Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Lin Wang
- Department of Otorhinolaryngology Head and Neck Surgery Binzhou People's Hospital Binzhou China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Lanjie Lei
- Institute of Translational Medicine Zhejiang Shuren University Hangzhou China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| |
Collapse
|
4
|
Diao Z, Li L, Zhou H, Yang L. Tannic acid and silicate-functionalized polyvinyl alcohol-hyaluronic acid hydrogel for infected diabetic wound healing. Regen Biomater 2024; 11:rbae053. [PMID: 38883183 PMCID: PMC11176089 DOI: 10.1093/rb/rbae053] [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: 01/24/2024] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 06/18/2024] Open
Abstract
Healing of chronic diabetic wounds is challenging due to complications of severe inflammatory microenvironment, bacterial infection and poor vascular formation. Herein, a novel injectable polyvinyl alcohol-hyaluronic acid-based composite hydrogel was developed, with tannic acid (TA) and silicate functionalization to fabricate an 'all-in-one' hydrogel PTKH. On one hand, after being locally injected into the wound site, the hydrogel underwent a gradual sol-gel transition in situ, forming an adhesive and protective dressing for the wound. Manipulations of rheological characteristics, mechanical properties and swelling ability of PTKH could be performed via regulating TA and silicate content in hydrogel. On the other hand, PTKH was capable of eliminating reactive oxygen species overexpression, combating infection and generating a cell-favored microenvironment for wound healing acceleration in vitro. Subsequent animal studies demonstrated that PTKH could greatly stimulate angiogenesis and epithelization, accompanied with inflammation and infection risk reduction. Therefore, in consideration of its impressive in vitro and in vivo outcomes, this 'all-in-one' multifunctional hydrogel may hold promise for chronic diabetic wound treatment.
Collapse
Affiliation(s)
- Zhentian Diao
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300131, China
| | - Longkang Li
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin 300131, China
| | - Huan Zhou
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300131, China
| | - Lei Yang
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300131, China
| |
Collapse
|
5
|
Zhou M, Lin X, Wang L, Yang C, Yu Y, Zhang Q. Preparation and Application of Hemostatic Hydrogels. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309485. [PMID: 38102098 DOI: 10.1002/smll.202309485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/28/2023] [Indexed: 12/17/2023]
Abstract
Hemorrhage remains a critical challenge in various medical settings, necessitating the development of advanced hemostatic materials. Hemostatic hydrogels have emerged as promising solutions to address uncontrolled bleeding due to their unique properties, including biocompatibility, tunable physical characteristics, and exceptional hemostatic capabilities. In this review, a comprehensive overview of the preparation and biomedical applications of hemostatic hydrogels is provided. Particularly, hemostatic hydrogels with various materials and forms are introduced. Additionally, the applications of hemostatic hydrogels in trauma management, surgical procedures, wound care, etc. are summarized. Finally, the limitations and future prospects of hemostatic hydrogels are discussed and evaluated. This review aims to highlight the biomedical applications of hydrogels in hemorrhage management and offer insights into the development of clinically relevant hemostatic materials.
Collapse
Affiliation(s)
- Minyu Zhou
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiang Lin
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Li Wang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Chaoyu Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yunru Yu
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Qingfei Zhang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
6
|
Cao H, Wang J, Hao Z, Zhao D. Gelatin-based biomaterials and gelatin as an additive for chronic wound repair. Front Pharmacol 2024; 15:1398939. [PMID: 38751781 PMCID: PMC11094280 DOI: 10.3389/fphar.2024.1398939] [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: 03/11/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Disturbing or disrupting the regular healing process of a skin wound may result in its progression to a chronic state. Chronic wounds often lead to increased infection because of their long healing time, malnutrition, and insufficient oxygen flow, subsequently affecting wound progression. Gelatin-the main structure of natural collagen-is widely used in biomedical fields because of its low cost, wide availability, biocompatibility, and degradability. However, gelatin may exhibit diverse tailored physical properties and poor antibacterial activity. Research on gelatin-based biomaterials has identified the challenges of improving gelatin's poor antibacterial properties and low mechanical properties. In chronic wounds, gelatin-based biomaterials can promote wound hemostasis, enhance peri-wound antibacterial and anti-inflammatory properties, and promote vascular and epithelial cell regeneration. In this article, we first introduce the natural process of wound healing. Second, we present the role of gelatin-based biomaterials and gelatin as an additive in wound healing. Finally, we present the future implications of gelatin-based biomaterials.
Collapse
Affiliation(s)
- Hongwei Cao
- Department of Otorhinolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jingren Wang
- Department of Prosthodontics, Affiliated Stomatological Hospital of China Medical University, Shenyang, China
| | - Zhanying Hao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Danyang Zhao
- Department of emergency Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Li L, Li H, Diao Z, Zhou H, Bai Y, Yang L. Development of a tannic acid- and silicate ion-functionalized PVA-starch composite hydrogel for in situ skeletal muscle repairing. J Mater Chem B 2024; 12:3917-3926. [PMID: 38536012 DOI: 10.1039/d3tb03006g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The repair capacity of skeletal muscle is severely diminished in massive skeletal muscle injuries accompanied by inflammation, resulting in muscle function loss and scar tissue formation. In the current work, we developed a tannic acid (TA)- and silicate ion-functionalized tissue adhesive poly(vinyl alcohol) (PVA)-starch composite hydrogel, referred to as PSTS (PVA-starch-TA-SiO32-). It was formed based on the hydrogen bonding of TA to organic polymers, as well as silicate-TA ligand interaction. PSTS could be gelatinized in minutes at room temperature with crosslinked network formation, making it applicable for injection. Further investigations revealed that PSTS had skeletal muscle-comparable conductivity and modulus to act as a temporary platform for muscle repairing. Moreover, PSTS could release TA and silicate ions in situ to inhibit bacterial growth, induce vascularization, and reduce oxidation, paving the way to the possibility of creating a favorable microenvironment for skeletal muscle regeneration and tissue fibrosis control. The in vivo model confirmed that PSTS could enhance muscle fiber regeneration and myotube formation, as well as reduce infection and inflammation risk. These findings thereby implied the great potential of PSTS in the treatment of formidable skeletal muscle injuries.
Collapse
Affiliation(s)
- Longkang Li
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300130, China
| | - Huipeng Li
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, China.
| | - Zhentian Diao
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300130, China
| | - Huan Zhou
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, China.
| | - Yanjie Bai
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, China.
- Department of Chemical Engineering, Hebei University of Technology, Tianjin, 300130, China.
| | - Lei Yang
- Center for Health Science and Engineering, Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, China.
| |
Collapse
|
8
|
Jiang F, Li L, Tian Y, Su Y, Zhao T, Ren R, Chi Z, Liu C. Enteromorpha Prolifera Polysaccharide-Derived Injectable Hydrogel for Fast Intraoperative Hemostasis and Accelerated Postsurgical Wound Healing Following Tumor Resection. Adv Healthc Mater 2024; 13:e2303456. [PMID: 38142288 DOI: 10.1002/adhm.202303456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/21/2023] [Indexed: 12/25/2023]
Abstract
Intraoperative bleeding and delayed postsurgical wound healing caused by persistent inflammation can increase the risk of tumor recurrence after surgical resection. To address these issues, Enteromorpha prolifera polysaccharide (PEP) with intrinsic potentials for hemostasis and wound healing, is chemically modified into aldehyde-PEP and hydrazine-PEP. Thereby, an injectable double-network hydrogel (OPAB) is developed via forming dual dynamic bonding of acylhydrazone bonds between the decorated aldehyde and hydrazine groups and hydrogen bonds between hydroxyl groups between boric acid and PEP skeletons. The OPAB exhibits controllable shape-adaptive gelation (35.0 s), suitable mechanical properties, nonstimulating self-healing (60 s), good wet tissue adhesion (30.9 kPa), and pH-responsive biodegradability. For in vivo models, owing to these properties, OPAB can achieve rapid hemostasis within 30 s for the liver hemorrhage, and readily loading of curcumin nanoparticles to remarkably accelerate surgical wound closure by alleviating inflammation, re-epithelialization, granulation tissue formation, and collagen deposition. Overall, this multifunctional injectable hydrogel is a promising material that facilitates simultaneous intraoperative hemorrhage and postsurgical wound repair, holding significant potential in the clinical managements of bleeding and surgical wounds for tumor resection.
Collapse
Affiliation(s)
- Fei Jiang
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China
| | - Luxi Li
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China
| | - Yu Tian
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China
| | - Yun Su
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China
| | - Tiange Zhao
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China
| | - Ruyi Ren
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China
| | - Zhe Chi
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China
| | - Chenguang Liu
- College of Marine Life Sciences, Ocean University of China, No. 5 Yushan Road, Qingdao, 266003, China
| |
Collapse
|
9
|
Li Y, Mu X, Feng W, Gao M, Wang Z, Bai X, Ren X, Lu Y, Zhou X. Supramolecular prodrug-like nanotheranostics with dynamic and activatable nature for synergistic photothermal immunotherapy of metastatic cancer. J Control Release 2024; 367:354-365. [PMID: 38286337 DOI: 10.1016/j.jconrel.2024.01.052] [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: 11/18/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 01/31/2024]
Abstract
Synergistic photothermal immunotherapy has attracted widespread attention due to the mutually reinforcing therapeutic effects on primary and metastatic tumors. However, the lack of clinical approval nanomedicines for spatial, temporal, and dosage control of drug co-administration underscores the challenges facing this field. Here, a photothermal agent (Cy7-TCF) and an immune checkpoint blocker (NLG919) are conjugated via disulfide bond to construct a tumor-specific small molecule prodrug (Cy7-TCF-SS-NLG), which self-assembles into prodrug-like nano-assemblies (PNAs) that are self-delivering and self-formulating. In tumor cells, over-produced GSH cleaves disulfide bonds to release Cy7-TCF-OH, which re-assembles into nanoparticles to enhance photothermal conversion while generate reactive oxygen species (ROSs) upon laser irradiation, and then binds to endogenous albumin to activate near-infrared fluorescence, enabling multimodal imaging-guided phototherapy for primary tumor ablation and subsequent release of tumor-associated antigens (TAAs). These TAAs, in combination with the co-released NLG919, effectively activated effector T cells and suppressed Tregs, thereby boosting antitumor immunity to prevent tumor metastasis. This work provides a simple yet effective strategy that integrates the supramolecular dynamics and reversibility with stimuli-responsive covalent bonding to design a simple small molecule with synergistic multimodal imaging-guided phototherapy and immunotherapy cascades for cancer treatment with high clinical value.
Collapse
Affiliation(s)
- Yajie Li
- College of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Xueluer Mu
- College of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Wenbi Feng
- College of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Min Gao
- College of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Zigeng Wang
- College of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Xue Bai
- College of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Xiangru Ren
- College of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China
| | - Yingxi Lu
- College of Material Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Xianfeng Zhou
- College of Material Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China; College of Polymer Science and Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| |
Collapse
|
10
|
Yang Y, Ye G, Qiu X. 3D sponge loaded with cisplatin-CS-calcium alginate MPs utilized as a void-filling prosthesis for the efficient postoperative prevention of tumor recurrence and metastasis. RSC Adv 2024; 14:7517-7527. [PMID: 38440275 PMCID: PMC10910265 DOI: 10.1039/d3ra07516h] [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: 11/03/2023] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
Intraoperative bleeding is a pivotal factor in the initiation of early recurrence and tumor metastasis following breast cancer excision. Distinct advantages are conferred upon postoperative breast cancer treatment through the utilization of locally administered implant therapies. This study devised a novel 3D sponge implant containing cisplatin-loaded chitosan-calcium alginate MPs capable of exerting combined chemotherapy and hemostasis effects. This innovative local drug-delivery implant absorbed blood and residual tumor cells post-tumor resection. Furthermore, the cisplatin-loaded chitosan-calcium alginate MPs sustainably targeted and eliminated cancer cells, thereby diminishing the risk of local recurrence and distant metastasis. This hydrogel material can also contribute to breast reconstruction, indicating the potential application of the 3D sponge in drug delivery for breast cancer treatment.
Collapse
Affiliation(s)
- Yihong Yang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University Guangzhou Guangdong 510515 P. R. China
| | - Genlan Ye
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University Guangzhou Guangdong 510515 P. R. China
| | - Xiaozhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University Guangzhou Guangdong 510515 P. R. China
| |
Collapse
|
11
|
Liu Y, He J, Li M, Ren K, Zhao Z. Inflammation-Driven Nanohitchhiker Enhances Postoperative Immunotherapy by Alleviating Prostaglandin E2-Mediated Immunosuppression. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6879-6893. [PMID: 38300288 DOI: 10.1021/acsami.3c17357] [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: 02/02/2024]
Abstract
Inflammation contributes to the immunosuppressive microenvironment and leads to the recurrence of surgically resected tumors. The COX-2/PGE2 axis is considered a key player in shaping the immunosuppression microenvironment. However, targeted modulation of the postoperative tumor microenvironment is challenging. To specifically curb the inflammation and alleviate immunosuppression, here, we developed a PGE2 inhibitor celecoxib (CXB)-loaded bionic nanoparticle (CP@CM) coated with activated murine vascular endothelial cell (C166 cells) membrane to target postoperative melanoma and inhibit its recurrence. CP@CM adhered to inflammatory white blood cells (WBCs) through the adhesion molecules, including ICAM-1, VCAM-1, E-selectin, and P-selection, expressed on the surface of C166 cells. Leveraging the natural tropism of the WBC to the inflammatory postoperative tumor site, CP@CM efficiently targeted postoperative tumors. In melanoma postoperative recurrence models, CXB significantly reduced PGE2 secretion and the recruitment of immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Treg) by inhibiting the activity of COX-2. This was followed by an increase in the infiltration of CD8+ T cells and CD4+ T cells in tumor tissues. Additionally, the immune responses were further enhanced by combining a PD-L1 monoclonal antibody. Ultimately, this immunotherapeutic strategy reversed the tumor immunosuppressive microenvironment and inhibited tumor recurrence, demonstrating a promising potential for postoperative immunotherapy for melanoma.
Collapse
Affiliation(s)
- Yingke Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan China
| | - Jiao He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Kebai Ren
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan China
| |
Collapse
|
12
|
Ebrahimnia M, Alavi S, Vaezi H, Karamat Iradmousa M, Haeri A. Exploring the vast potentials and probable limitations of novel and nanostructured implantable drug delivery systems for cancer treatment. EXCLI JOURNAL 2024; 23:143-179. [PMID: 38487087 PMCID: PMC10938236 DOI: 10.17179/excli2023-6747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/08/2024] [Indexed: 03/17/2024]
Abstract
Conventional cancer chemotherapy regimens, albeit successful to some extent, suffer from some significant drawbacks, such as high-dose requirements, limited bioavailability, low therapeutic indices, emergence of multiple drug resistance, off-target distribution, and adverse effects. The main goal of developing implantable drug delivery systems (IDDS) is to address these challenges and maintain anti-cancer drugs directly at the intended sites of therapeutic action while minimizing inevitable side effects. IDDS possess numerous advantages over conventional drug delivery, including controlled drug release patterns, one-time drug administration, as well as loading and stabilizing poorly water-soluble chemotherapy drugs. Here, we summarized conventional and novel (three-dimensional (3D) printing and microfluidic) preparation techniques of different IDDS, including nanofibers, films, hydrogels, wafers, sponges, and osmotic pumps. These systems could be designed with high biocompatibility and biodegradability features using a wide variety of natural and synthetic polymers. We also reviewed the published data on these systems in cancer therapy with a particular focus on their release behavior. Various release profiles could be attained in IDDS, which enable predictable, adjustable, and sustained drug releases. Furthermore, multi-step or stimuli-responsive drug release could be obtained in these systems. The studies mentioned in this article have proven the effectiveness of IDDS for treating different cancer types with high prevalence, including breast cancer, and aggressive cancer types, such as glioblastoma and liver cancer. Additionally, the challenges in applying IDDS for efficacious cancer therapy and their potential future developments are also discussed. Considering the high potential of IDDS for further advancements, such as programmable release and degradation features, further clinical trials are needed to ensure their efficiency. The overall goal of this review is to expand our understanding of the behavior of commonly investigated IDDS and to identify the barriers that should be addressed in the pursuit of more efficient therapies for cancer. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Maryam Ebrahimnia
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sonia Alavi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Hamed Vaezi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdieh Karamat Iradmousa
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Hu Y, Wang H, Liu Y. NETosis: Sculpting tumor metastasis and immunotherapy. Immunol Rev 2024; 321:263-279. [PMID: 37712361 DOI: 10.1111/imr.13277] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
The process of neutrophil extracellular traps (NETs) formation, called NETosis, is a peculiar death modality of neutrophils, which was first observed as an immune response against bacterial infection. However, recent work has revealed the unique biology of NETosis in facilitating tumor metastatic process. Neutrophil extracellular traps released by the tumor microenvironment (TME) shield tumor cells from cytotoxic immunity, leading to impaired tumor clearance. Besides, tumor cells tapped by NETs enable to travel through vessels and subsequently seed distant organs. Targeted ablation of NETosis has been proven to be beneficial in potentiating the efficacy of cancer immunotherapy in the metastatic settings. This review outlines the impact of NETosis at almost all stages of tumor metastasis. Furthermore, understanding the multifaceted interplay between NETosis and the TME components is crucial for supporting the rational development of highly effective combination immunotherapeutic strategies with anti-NETosis for patients with metastatic disease.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Houhong Wang
- Department of General Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, China
| | - Yang Liu
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
14
|
Wang X, Yuan K, Su Y, Li X, Meng L, Zhao N, Hu Y, Duan F, Xu FJ. Tuning Blood-Material Interactions to Generate Versatile Hemostatic Powders and Gels. Adv Healthc Mater 2024; 13:e2301945. [PMID: 37897223 DOI: 10.1002/adhm.202301945] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/05/2023] [Indexed: 10/29/2023]
Abstract
Polymer-based hemostatic materials/devices have been increasingly exploited for versatile clinical scenarios, while there is an urgent need to reveal the rational design/facile approach for procoagulant surfaces through regulating blood-material interactions. In this work, degradable powders (PLPS) and thermoresponsive gels (F127-PLPS) are readily developed as promising hemostatic materials for versatile clinical applications, through tuning blood-material interactions with optimized grafting of cationic polylysine: the former is facilely prepared by conjugating polylysine onto porous starch particle, while F127-PLPS is prepared by the simple mixture of PLPS and commercial thermosensitive polymer. In vitro and in vivo results demonstrate that PLPS2 with the optimal-/medium content of polylysine grafts achieve the superior hemostatic performance. The underlying procoagulant mechanism of PLPS2 surface is revealed as the selective fibrinogen adsorption among the competitive plasma-protein-adsorption process, which is the foundation of other blood-material interactions. Moreover, in vitro results confirm the achieved procoagulant surface of F127-PLPS through optimal PLPS2 loading. Together with the tunable thermoresponsiveness, F127-PLPS exhibits outstanding hemostatic utilization in both femoral-artery-injury and renal-artery-embolization models. The work thereby pioneers an appealing approach for generating versatile polymer-based hemostatic materials/devices.
Collapse
Affiliation(s)
- Xueru Wang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kai Yuan
- Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Su
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoyue Li
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Limin Meng
- Department of Medical Imaging, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Nana Zhao
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yang Hu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Feng Duan
- Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
15
|
Luo Y, Tao F, Wang J, Chai Y, Ren C, Wang Y, Wu T, Chen Z. Development and evaluation of tilapia skin-derived gelatin, collagen, and acellular dermal matrix for potential use as hemostatic sponges. Int J Biol Macromol 2023; 253:127014. [PMID: 37742900 DOI: 10.1016/j.ijbiomac.2023.127014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/15/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
Hemostasis plays a critical role in the early stage of wound healing, especially in acute wounds which can significantly improve the survival of patients. Based on the excellent biocompatibility of natural biomaterials, in this study, we prepared a series of novel hemostatic sponges by using tilapia skin, a marine biological resource, and extracting tilapia skin-derived gelatin, collagen, and acellular dermal matrix through five different methods. Using in vitro sheep blood and in vivo rat liver hemorrhage models, we found that tilapia skin sponges had excellent coagulation and hemostatic abilities. Among them, the collagen sponge exhibited optimal hemostasis performance because it could accelerate clotting by binding to the specific sites of blood cells and platelets. Furthermore, the sponges' porous structure enhanced the capability to absorb blood, thus effectively promoting hemostasis. In summary, we reported an efficient and convenient method to prepare marine biological resources into sponges, which provided a novel class of alternatives for hemostasis in acute wounds with broad application prospects.
Collapse
Affiliation(s)
- Yanan Luo
- Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Fulin Tao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
| | - Jing Wang
- Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, College of Textile & Clothing, Qingdao University, Qingdao 266071, China
| | - Yandong Chai
- Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Chaohua Ren
- Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Yuanfei Wang
- Qingdao Medical College, Qingdao University, Qingdao 266071, China; Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, College of Textile & Clothing, Qingdao University, Qingdao 266071, China.
| | - Tong Wu
- Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266071, China; Qingdao Medical College, Qingdao University, Qingdao 266071, China; Shandong Key Laboratory of Medical and Health Textile Materials, Collaborative Innovation Center for Eco-textiles of Shandong Province and the Ministry of Education, College of Textile & Clothing, Qingdao University, Qingdao 266071, China.
| | - Zhenyu Chen
- Department of Cosmetic and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao 266071, China.
| |
Collapse
|
16
|
Sang F, Pan L, Ji Z, Zhang B, Meng Z, Cao L, Zhang J, Li X, Yang X, Shi C. Polydopamine functionalized polyurethane shape memory sponge with controllable expansion performance triggered by near-infrared light for incompressible hemorrhage control. Colloids Surf B Biointerfaces 2023; 232:113590. [PMID: 37862950 DOI: 10.1016/j.colsurfb.2023.113590] [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: 01/05/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/22/2023]
Abstract
Uncontrolled expansion of shape memory sponges face a significant challenge in the treatment of lethal incompressible hemorrhage, which can lead to blood overflow or damage to the surrounding tissue. Herein, we developed a polydopamine functionalized polyurethane shape memory sponge (PDA-TPI-PU) with a controllable degree of expansion by near-infrared (NIR) light-triggered stimulation for the treatment of incompressible hemorrhage. The sponge has excellent liquid absorption performance and robust mechanical strength as well as good photothermal conversion ability. Under NIR light of 0.32 W/cm2, the maximum recovery rate of the fixed-shape compression sponge was 91% within 25 s in air and 80% within 25 s in blood. In the SD rat liver penetrating injury model, compared with commercial medical gelatin sponge and PVA sponge, the PDA-TPI-PU sponge could effectively control the bleeding under the NIR light irradiation and did not cause excessive compression of the wound. The sponge with these characteristics shows potential application prospects as a hemostatic material.
Collapse
Affiliation(s)
- Feng Sang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| | - Luqi Pan
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| | - Zhixiao Ji
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| | - Bingxu Zhang
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| | - Zhizhen Meng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Lina Cao
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| | - Jing Zhang
- College of Materials Science and Engineering, Donghua University, Shanghai 200051, China
| | - Xujian Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China.
| | - Xiao Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China.
| | - Changcan Shi
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
17
|
Li N, Zhang G, Liu Y, Sun L, Zhao X, Ding L, Liu Y, Wang M, Ren X. A Natural Self-Assembled Gel-Sponge with Hierarchical Porous Structure for Rapid Hemostasis and Antibacterial. Adv Healthc Mater 2023; 12:e2301465. [PMID: 37449760 DOI: 10.1002/adhm.202301465] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Developing hemostatic agents with reliable biosafety and high efficiency has paramount clinical significance for saving lives. Herein, inspired from traditional Chinese medicine, a sponge (BC-S) with hierarchical porous structure is proposed for the treatment of bleeding. The BC-S is prepared by a simple self-assembly method employing Bletilla Striata polysaccharide and quaternary amine alkaloids (QA) from Bletilla Striata and Coptidis Rhizoma. The ideal cation donor encapsulated in the helical structure of BSP enlarges the inter-layer space of sponge by the action of electrostatic repulsion, forming wider channels which can accelerate the diversion speed of absorbed blood. Then, platelets and erythrocytes are trapped tightly in the reticular structure and extruded to deformation, activation. Subsequently, fibrin network forms and reinforces the internal multilayer mesh, blocks the outflow of blood. QA is released from the sponge skeleton mainly driven by a combination of surface erosion and potentially solution diffusion among pore to provide long-term antibacterial activity. Benefiting from the well-designed structure and the effective hemostatic mechanism, the BC-S displays more excellent hemostatic performance in different models in vivo and in vitro compared with typical gelatin hemostatic sponge. This work is expected to boost the development of emerging hemostatic agents.
Collapse
Affiliation(s)
- Na Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Guoqin Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yi Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lili Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xin Zhao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Liqin Ding
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yanan Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Meng Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| |
Collapse
|
18
|
Liu H, Zhu X, Nie L, Guo L, Jiang C, Wang G, Huang W, Hou L, Hu T, Yakovlev AN, Xu X, Yu X, Wang T. Multimode-Responsive Luminescence of Er 3+ Single-Activated CaF 2 Phosphor for Advanced Information Encryption. Inorg Chem 2023; 62:16485-16492. [PMID: 37738045 DOI: 10.1021/acs.inorgchem.3c02215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
The current optical anticounterfeit strategies that rely on multimode luminescence in response to the photon or thermal stimuli have significant importance in the field of anticounterfeiting and information encryption. However, the dependence on light and heat sources might limit their flexibility in practical applications. In this work, Er3+ single-doped CaF2 phosphors that show multistimuli-responsive luminescence have been successfully prepared. The as-obtained CaF2:Er3+ phosphor exhibits green photoluminescence (PL) and color-tunable up-conversation (UC) luminescence from red to green due to the cross-relaxation of Er3+ ions. Additionally, as-obtained CaF2:Er3+ phosphors also display green mechano-luminescence behavior, which is induced by the contact electrification between the CaF2 particles and PDMS polymers, enabling the phosphor to flexibly respond to mechanical stimuli. Moreover, feasible anticounterfeiting schemes with the capability of multistimuli-responsive and flexible decryption have been constructed, further expanding the application of optical materials in the field of advanced anticounterfeiting and information encryption.
Collapse
Affiliation(s)
- Haozhe Liu
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Xuanyu Zhu
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Lin Nie
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Longchao Guo
- School of Mechanical Engineering, Institute for Advanced Materials, Chengdu University, Chengdu 610106, China
| | - Chaoxin Jiang
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Guohao Wang
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Wenlong Huang
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Lihui Hou
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Tingting Hu
- T.F. Gorbachev Kuzbass State Technical University, 28, Vesennyaya Street, Kemerovo 650000, Russia
| | | | - Xuhui Xu
- Faculty of Materials Science and Engineering, Key Laboratory of Advanced Materials of Yunnan Province, Kunming University of Science and Technology, Kunming 650093, China
| | - Xue Yu
- School of Mechanical Engineering, Institute for Advanced Materials, Chengdu University, Chengdu 610106, China
| | - Ting Wang
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| |
Collapse
|
19
|
Kuang G, Zhang Q, Yu Y, Shang L, Zhao Y. Cryo-shocked cancer cell microgels for tumor postoperative combination immunotherapy and tissue regeneration. Bioact Mater 2023; 28:326-336. [PMID: 37346097 PMCID: PMC10279695 DOI: 10.1016/j.bioactmat.2023.05.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/08/2023] [Accepted: 05/30/2023] [Indexed: 06/23/2023] Open
Abstract
Prevention of recurrence/metastasis and tissue regeneration are critical for post-surgery treatment of malignant tumors. Here, to address these needs, a novel type of microgel co-loading cryo-shocked cancer cells, immunoadjuvant, and immune checkpoint inhibitor is presented by microfluidic electrospray technology and liquid nitrogen treatment. Owing to the encapsulation of cryo-shocked cancer cells and immunoadjuvant, the microgels can recruit dendritic cells and activate them in situ, and evoke a robust immune response. Moreover, with the combination of the immune checkpoint inhibitor, the antitumor immune response is further enhanced by inhibiting the interaction of PD1 and PDL1. With this, the excellent anti-recurrence and anti-metastasis efficacy of the microgels are demonstrated in an orthotopic breast cancer mouse model. Besides, because of the excellent biocompatibility and appropriate degradation performance, the microgels can provide support for normal cell adhesion and growth, which is beneficial to tissue reconstruction. These properties indicate the great value of the cryo-shocked cancer cell microgels for efficient tumor postoperative combination immunotherapy and tissue regeneration.
Collapse
Affiliation(s)
- Gaizhen Kuang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Qingfei Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yunru Yu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Luoran Shang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, The International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
20
|
Wang P, Yang Y, Wen H, Li D, Zhang H, Wang Y. Progress in construction and release of natural polysaccharide-platinum nanomedicines: A review. Int J Biol Macromol 2023; 250:126143. [PMID: 37544564 DOI: 10.1016/j.ijbiomac.2023.126143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Natural polysaccharides are natural biomaterials that have become candidate materials for nano-drug delivery systems due to their excellent biodegradability and biocompatibility. Platinum (Pt) drugs have been widely used in the clinical therapy for various solid tumors. However, their extensive systemic toxicity and the drug resistance acquired by cancer cells limit the applications of platinum drugs. Modern nanobiotechnology provides the possibility for targeted delivery of platinum drugs to the tumor site, thereby minimizing toxicity and optimizing the efficacies of the drugs. In recent years, numerous natural polysaccharide-platinum nanomedicine delivery carriers have been developed, such as nanomicelles, nanospheres, nanogels, etc. Herein, we provide an overview on the construction and drug release of natural polysaccharide-Pt nanomedicines in recent years. Current challenges and future prospectives in this field are also put forward. In general, combining with irradiation and tumor microenvironment provides a significant research direction for the construction of natural polysaccharide-platinum nanomedicines and the release of responsive drugs in the future.
Collapse
Affiliation(s)
- Pengge Wang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China; College of Biological and Pharmaceutical Engineering, Nanjing University of Technology, Nanjing City, Jiangsu Province 211816, China
| | - Yunxia Yang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China; Jiangsu Province Engineering Research Center of Agricultural Breeding Pollution Control and Resource, Yancheng Teachers University, Yancheng 224007, China; Jiangsu Key Laboratory for Bioresources of Saline Soils, Yancheng Teachers University, Yancheng 224007, China.
| | - Haoyu Wen
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Dongqing Li
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Hongmei Zhang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China
| | - Yanqing Wang
- School of Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng 224007, China.
| |
Collapse
|
21
|
Chen XJ, Lei ZY, Liu P, Lei MJ, Xu H, Yu LJ, Ao MZ. An aminocaproic acid-grafted chitosan derivative with superior antibacterial and hemostatic properties for the prevention of secondary bleeding. Carbohydr Polym 2023; 316:120988. [PMID: 37321717 DOI: 10.1016/j.carbpol.2023.120988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/19/2023] [Accepted: 05/04/2023] [Indexed: 06/17/2023]
Abstract
Uncontrolled bleeding is one of the leading causes of human mortality. Existing hemostatic materials or techniques cannot meet the clinical requirements for safe and effective hemostasis. The development of novel hemostatic materials has always been of great interest. Chitosan hydrochloride (CSH), a derivative of chitin, is extensively used on wounds as an antibacterial and hemostatic agent. However, the formation of intra- or intermolecular hydrogen bonds between hydroxyl and amino groups limits its water solubility and dissolution rate and affects its effectiveness in promoting coagulation. Herein, we covalently grafted aminocaproic acid (AA) to the hydroxyl and amino groups of CSH via ester and amide bonds, respectively. The solubility of CSH in water (25 °C) was 11.39 ± 0.98 % (w/v), whereas the AA-grafted CSH (CSH-AA) reached 32.34 ± 1.23 % (w/v). Moreover, the dissolution rate of CSH-AA in water was 6.46 times higher than that of CSH. Subsequent studies proved that CSH-AA is non-toxic, biodegradable, and has superior antibacterial and hemostatic properties to CSH. Additionally, anti-plasmin activity can be exerted by the dissociated AA from the CSH-AA backbone, which can help to lessen secondary bleeding.
Collapse
Affiliation(s)
- Xiao-Juan Chen
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhi-Yong Lei
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Pan Liu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Meng-Jie Lei
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hang Xu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Long-Jiang Yu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics, Ministry of Education, Wuhan 430074, China; Hubei Engineering Research Center for both Edible and Medicinal Resources, Wuhan 430074, China.
| | - Ming-Zhang Ao
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics, Ministry of Education, Wuhan 430074, China; Hubei Engineering Research Center for both Edible and Medicinal Resources, Wuhan 430074, China.
| |
Collapse
|
22
|
Xu X, Wu Q, Tan L, Men X, Huang Y, Li H. Biomimetic Metal-Chalcogenide Agents Enable Synergistic Cancer Therapy via Microwave Thermal-Dynamic Therapy and Immune Cell Activation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:42182-42195. [PMID: 37651685 DOI: 10.1021/acsami.3c05728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Microwave thermal dynamic therapy (MTDT), which combines thermal effects and reactive oxygen species (ROS) by microwave activation, seems to be a promising anticancer therapeutic method. A multifunctional agent for achieving synergistic localized cancer treatment is the key to exploit the strategy to inhibit tumor cell recurrence and metastasis. In the study, a ZIF-67 based theranostic agent loaded with metal-chalcogenide open framework 3 (MCOF3) and heat shock protein 70 (HSP70) as the inner component was studied, coupled with targeting cancer cell membrane (TCM) as the biomimetic outer shell. We found that metal ions in MCOF3 enabled the composite agents to show peroxide-like activity to produce •OH and destroy cancer cells. And then, the microwave (MW) thermal sensitizer of ZIF-67 was used to specifically convert the MW energy into thermal energy and selectively heat the tumor via the cell's targeting. Additionally, the effect of continuous MW thermal therapy has been shown to promote the expression of HSP70, and further activate the effector of CD4 T cell and CD8α T cell. As such, the agents effectively inhibit the tumor cell growth under MW irradiation in vitro and in vivo due to the synergistic effects of MTDT and immune cell activation. The study provides an emerging strategy to ablation cancer effectively.
Collapse
Affiliation(s)
- Xiaomu Xu
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China
- Laboratory of Controllable Preparation and Application of Nanomaterials, Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, China
| | - Qiong Wu
- Laboratory of Controllable Preparation and Application of Nanomaterials, Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Longfei Tan
- Laboratory of Controllable Preparation and Application of Nanomaterials, Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xianwei Men
- Laboratory of Controllable Preparation and Application of Nanomaterials, Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yue Huang
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, China
- School of Stomatology, Jinan University, Guangzhou 510632, China
| | - Hong Li
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 511436, China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou 510632, China
| |
Collapse
|
23
|
Zhong T, Yu J, Pan Y, Zhang N, Qi Y, Huang Y. Recent Advances of Platinum-Based Anticancer Complexes in Combinational Multimodal Therapy. Adv Healthc Mater 2023; 12:e2300253. [PMID: 37097737 DOI: 10.1002/adhm.202300253] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/19/2023] [Indexed: 04/26/2023]
Abstract
Platinum drugs with manifest therapeutic effects are widely used, but their systemic toxicity and the drug resistance acquired by cancer cells limit their clinical applications. Thus, the exploration on appropriate methods and strategies to overcome the limitations of traditional platinum drugs becomes extremely necessary. Combination therapy of platinum drugs can inhibit tumor growth and metastasis in an additive or synergistic manner, and can potentially reduce the systemic toxicity of platinum drugs and overcome platinum-resistance. This review summarizes the various modalities and current progress in platinum-based combination therapy. The synthetic strategies and therapeutic effects of some platinum-based anticancer complexes in the combination of platinum drugs with gene editing, ROS-based therapy, thermal therapy, immunotherapy, biological modelling, photoactivation, supramolecular self-assembly and imaging modality are briefly described. Their potential challenges and prospects are also discussed. It is hoped that this review will inspire researchers to have more ideas for the future development of highly effective platinum-based anti-cancer complexes.
Collapse
Affiliation(s)
- Tianyuan Zhong
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| | - Jie Yu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| | - Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| | - Ning Zhang
- The Second Affiliated Hospital of Harbin Medical University, Department of Orthopedics, Harbin, 150000, China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
- Key Laboratory of Sustainable Advanced Functional Materials of Jilin Province, Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
24
|
Chen Y, Guo J, Wu X, Xu Y, Wang J, Ren H, Zhao Y. Microfluidic spinning of natural origin microfibers for breast tumor postsurgical treatment. CHEMICAL ENGINEERING JOURNAL 2023; 472:144901. [DOI: 10.1016/j.cej.2023.144901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
|
25
|
Lan JS, Zeng RF, Li Z, Wu Y, Liu L, Chen LX, Liu Y, He YT, Zhang T, Ding Y. CD44-Targeted Photoactivatable Polymeric Nanosystem with On-Demand Drug Release as a "Photoactivatable Bomb" for Combined Photodynamic Therapy-Chemotherapy of Cancer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:34554-34569. [PMID: 37462246 DOI: 10.1021/acsami.3c05645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Nowadays, the combined use of chemotherapy and photodynamic therapy (PDT) remains the most popular strategy for cancer treatment with high theraprutic efficacy. However, targeted therapy with the on-demand release of drugs is what most clinical treatments lack, leading to heavy side effects. Herein, a new CD44-targeted and red-light-activatable nanosystem, Ru-HA@DOX nanoparticles (NPs), was developed by conjugating hydrophilic biodegradable hyaluronic acid (HA) and hydrophobic photoresponsive ruthenium (Ru) complexes, which could encapsulate the chemotherapeutic drug doxrubicin (DOX). Ru-HA@DOX NPs can selectively accumulate at the tumor through the enhanced permeability and retention (EPR) effect and CD44-mediated endocytosis, thus avoiding off-target toxicity during circulation. After 660 nm of irradiation at the tumor site, Ru-HA@DOX NPs, as a "photoactivatable bomb", was split via the photocleavable Ru-N coordination bond to fast release DOX and produce singlet oxygen (1O2) for PDT. In general, Ru-HA@DOX NPs retained its integrity before irradiation and possessed minimal cytotoxicity, while under red-light irradiation, Ru-HA@DOX NPs showed significant cytotoxicity due to the release of DOX and production of 1O2 at the tumor. Chemotherapy-PDT of Ru-HA@DOX NPs resulted in a significant inhibition of tumor growth in A549-tumor-bearing mice and reduced the cardiotoxicity of DOX. Therefore, this study offers a novel CD44-targeted drug-delivery system with on-demand drug release for synergistic chemotherapy-PDT.
Collapse
Affiliation(s)
- Jin-Shuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rui-Feng Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ya Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li-Xia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yun Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Tian He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
26
|
Zhang M, Xu W, Li X, Ling G, Zhang P. Tunicate-mimetic antibacterial hydrogel based on metal ion crosslinking and chitosan functionalization for wound healing. Int J Biol Macromol 2023:125062. [PMID: 37247717 DOI: 10.1016/j.ijbiomac.2023.125062] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/13/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
With the increasing prevalence of drug-resistant bacterial infections and frequent occurrences of slow wound healing, the development of novel antibacterial wound dressings has become a serious challenge. Hydrogel dressings have attracted extensive attention on wound healing due to their unique three-dimensional network structures and properties. However, it is a challenge to develop natural long-acting antibacterial hydrogels with multiple functions such as excellent cell affinity, wet adhesion and mechanical properties. Inspired by the wound healing mechanism and adhesion characteristics of tunicates, a series of biomimetic antibacterial hydrogels were prepared by utilizing pyrogallol-modified chitosan (GACS) and polyvinyl alcohol (PVA) as matrix, zinc ions (Zn2+) as crosslinking and antibacterial agents, and ethyl N-lauroyl l-arginate hydrochloride (LAE) as the antibacterial active ingredient. The morphology, swelling, water retention, degradability, wet adhesion, biocompatibility, mechanical and rheological properties of PVA/GACS/Zn2+/LAE hydrogels were evaluated. And the adhesion ability conferred by the pyrogallol structures enabled the hydrogel with enhanced antibacterial effect and hemostatic ability. Moreover, the in vivo experiments on rat models with full-thickness infected wounds confirmed that PVA/GACS/Zn2+/LAE hydrogels could efficiently kill bacteria, significantly improve the wound microenvironment, greatly promote fibroblast proliferation and collagen deposition and ultimately accelerate wound healing. In a word, this study provided a feasible and simple way for the development of biomimetic antibacterial hydrogel dressings applied in infected wounds, which could not only seal wounds with various shapes and provide a moist and antibacterial environment for wounds, but also have certain mechanical strength, excellent wound adhesion, good biocompatibility and hemostatic performance.
Collapse
Affiliation(s)
- Manyue Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Wenxin Xu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Xiaodan Li
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Guixia Ling
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Peng Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
27
|
Interpenetrating network expansion sponge based on chitosan and plasma for ultrafast hemostasis of arterial bleeding wounds. Carbohydr Polym 2023; 307:120590. [PMID: 36781269 DOI: 10.1016/j.carbpol.2023.120590] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/27/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023]
Abstract
Preventing arterial hemorrhage by intervening within the first few minutes is critical to the patient's life. Hemostatic materials have been developed over the last decades to address this issue, nevertheless these materials alone do not contribute to improve the survival effects in many extreme conditions, which is usually caused by penetrating arterial bleeding wounds that are incompressible and deep arterial bleeding with irregularly shapes. It is well known that, after calcium ion stimulation, many intriguing changes occurred in the major components of plasma, including the activation of several coagulation factors, such as the conversion of fibrinogen to fibrin, prothrombin to thrombin, and so on. Therefore, we constructed an expansion sponge with interpenetrating network based on chitosan and plasma, while various activated coagulation factors in plasma were also loaded into the pore structure of chitosan sponges. The prepared CS-PG sponge is capable of providing a simpler and more efficient method for treating high-pressure arterial bleeding wounds, which includes three steps: Rapid sealing and adhension, Thrombin catalysis and Activated autocoagulation. As the next generation bioactive materials, compared to conventional hemostatic materials, CS-PG sponge demonstrated superior hemostatic characteristics in both rabbit femoral artery damage and rat liver injury models.
Collapse
|
28
|
Chen J, Zhao L, Ling J, Yang LY, Ouyang XK. A quaternized chitosan and carboxylated cellulose nanofiber-based sponge with a microchannel structure for rapid hemostasis and wound healing. Int J Biol Macromol 2023; 233:123631. [PMID: 36775224 DOI: 10.1016/j.ijbiomac.2023.123631] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/13/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
Abstract
A hemostatic sponge should perform rapid hemostasis and exhibit antibacterial properties, whilst being non-toxic, breathable, and degradable. This study prepared a hemostatic sponge (CQTC) with microchannels, specifically a microchannel structure based on quaternized chitosan (QCS) and carboxylated cellulose nanofibers (CCNF) obtained by using tannic acid and Cu2+ complex (crosslinking agent). The sponge had low density and high porosity, while being degradable. The combination of microchannels and three-dimensional porous structure of CQTC leads to excellent liquid absorption and hemostasis ability, based on a liquid absorption rate test and in vitro hemostasis experiment. In addition, CQTC exhibited excellent antibacterial activity against both gram-negative and gram-positive bacteria, and it promoted wound healing. In conclusion, this porous and microchannel hemostatic sponge has broad application prospects as a clinical wound hemostatic material.
Collapse
Affiliation(s)
- Jing Chen
- Zhoushan Maternal and Child Care Hospital, Zhoushan 316000, PR China
| | - Lijuan Zhao
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, PR China
| | - Junhong Ling
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, PR China
| | - Li-Ye Yang
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, PR China
| | - Xiao-Kun Ouyang
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, PR China.
| |
Collapse
|
29
|
Zhao M, Xu R, Yang Y, Tong L, Liang J, Jiang Q, Fan Y, Zhang X, Sun Y. Bioabsorbable nano-micelle hybridized hydrogel scaffold prevents postoperative melanoma recurrence. J Control Release 2023; 356:219-231. [PMID: 36889462 DOI: 10.1016/j.jconrel.2023.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/10/2023]
Abstract
The residual and scattered small tumor tissue or cells after surgery are the main reason for tumor recurrence. Chemotherapy has a powerful ability to eradicate tumors but always accompanied by serious side effects. In this work, tissue-affinity mercapto gelatin (GelS) and dopamine-modified hyaluronic acid (HAD) were employed to fabricate a hybridized cross-linked hydrogel scaffold (HG) by multiple chemical reactions, which could integrate the doxorubicin (DOX) loaded reduction-responsive nano-micelle (PP/DOX) into this scaffold via click reaction to obtain the bioabsorbable nano-micelle hybridized hydrogel scaffold (HGMP). With the degradation of HGMP, PP/DOX was slowly released and formed targeted PP/DOX with degraded gelatin fragments as target molecules, which increased the intracellular accumulation, and inhibited the aggregation of B16F10 cells in vitro. In mouse models, HGMP absorbed the scattered B16F10 cells and released targeted PP/DOX to suppress tumorigenesis. For another, implantation of HGMP at the surgical site reduced the recurrence rate of postoperative melanoma and inhibited the growth of recurrent tumors. Meanwhile, HGMP significantly relieved the damage of free DOX to hair follicle tissue. This bioabsorbable nano-micelle hybridized hydrogel scaffold provided a valuable strategy for adjuvant therapy after tumor surgery.
Collapse
Affiliation(s)
- Mingda Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Ruiling Xu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Yuedi Yang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Lei Tong
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; Sichuan Testing Centre for Biomaterials and Medical Devices, No.29 Wangjiang Road, Chengdu, Sichuan 610064, PR China
| | - Qing Jiang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China; College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, PR China.
| |
Collapse
|
30
|
Wang H, Cheng J, Sun F, Dou X, Liu J, Wang Y, Li M, Gao J, Liu X, Wang X, Yang F, Zhu Z, Shen H, Zhang L, Tang P, Wu D. A Super Tough, Rapidly Biodegradable, Ultrafast Hemostatic Bioglue. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208622. [PMID: 36579739 DOI: 10.1002/adma.202208622] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Death happening due to massive hemorrhage has been involved in military conflicts, traffic accidents, and surgical injuries of various human disasters. Achieving rapid and effective hemostasis to save lives is crucial in urgent massive bleeding situations. Herein, a covalent cross-linked AG-PEG glue based on extracellular matrix-like amino-gelatin (AG) and PEG derivatives is developed. The AG-PEG glue gelatinizes fast and exhibits firm and indiscriminate close adhesion with various moist tissues upon being dosed. The formed glue establishes an adhesive and robust barrier to seal the arterial, hepatic, and cardiac hemorrhagic wounds, enabling it to withstand up to 380 mmHg blood pressure in comparison with normal systolic blood pressure of 60-180 mmHg. Remarkably, massive bleeding from a pig cardiac penetrating hole with 6 mm diameter is effectively stopped using the glue within 60 s. Postoperative indexes of the treated pig gradually recover and the cardiac wounds regrow significantly at 14 days. Possessing on-demand solubility, self-gelling, and rapid degradability, the AG-PEG glue may provide a fascinating stop-bleeding approach for clinical hemostasis and emergency rescue.
Collapse
Affiliation(s)
- Hufei Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junyao Cheng
- Senior Department of Orthopedics, National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Chinese PLA General Hospital, Beijing, 100853, China
| | - Feifei Sun
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Xueyu Dou
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
| | - Jianheng Liu
- Senior Department of Orthopedics, National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yiru Wang
- Department of Ultrasound, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Ming Li
- Senior Department of Orthopedics, National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianpeng Gao
- Senior Department of Orthopedics, National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiao Liu
- Senior Department of Orthopedics, National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei Yang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ziran Zhu
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Shen
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Licheng Zhang
- Senior Department of Orthopedics, National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Chinese PLA General Hospital, Beijing, 100853, China
| | - Peifu Tang
- Senior Department of Orthopedics, National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Chinese PLA General Hospital, Beijing, 100853, China
| | - Decheng Wu
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
31
|
Sun Y, Miao T, Wang Y, Wang X, Lin J, Zhao N, Hu Y, Xu FJ. A natural polyphenol-functionalized chitosan/gelatin sponge for accelerating hemostasis and infected wound healing. Biomater Sci 2023; 11:2405-2418. [PMID: 36799455 DOI: 10.1039/d2bm02049a] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Natural polymers have been particularly appealing for constructing hemostatic materials/devices, but it is still desirable to develop new natural polymer-based biomaterials with balanced hemostatic and wound-healing performance. In this work, a natural polyphenol-functionalized chitosan/gelatin sponge (PCGS) was prepared by the lyophilization of a chitosan/gelatin mixture solution (under a self-foaming condition to prepare the CGS) and subsequent chemical cross-linking with procyanidin (PC). Compared with the original CGS, PCGS exhibited an enhanced liquid-absorption ability, reduced surface charges, and similar/low hemolysis rate. Benefiting from such a liquid-absorption ability (∼4000% for whole blood and normal saline) and moderate surface charges, PCGS exhibited high in vitro hemostatic property and promising hemostatic performance in an in vivo femoral-artery-injury model. In addition, PCGS possessed higher antioxidant property and slightly decreased antibacterial ability than CGS, owing to the incorporation of PC. The feasibility of PCGS for treating infected wounds was further confirmed in an in vivo infected-tooth-extraction model, as the typical complication of intractable tooth-extraction bleeding. The present work demonstrated a facile approach for developing multifunctional hemostatic materials through the flexible management of natural polymers and polyphenols.
Collapse
Affiliation(s)
- Yujie Sun
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Tengfei Miao
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China. .,College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Yu Wang
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China. .,College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Xiaochen Wang
- Shandong Center for Food and Drug Evaluation & Inspection, Jinan 250014, China
| | - Jie Lin
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China. .,College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Nana Zhao
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China. .,College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Yang Hu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China. .,College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Fu-Jian Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China. .,College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| |
Collapse
|
32
|
Jing P, Luo Y, Chen Y, Tan J, Liao C, Zhang S. Aspirin-Loaded Cross-Linked Lipoic Acid Nanodrug Prevents Postoperative Tumor Recurrence by Residual Cancer Cell Killing and Inflammatory Microenvironment Improvement. Bioconjug Chem 2023; 34:366-376. [PMID: 36626242 DOI: 10.1021/acs.bioconjchem.2c00548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In addition to residual cancer cells, the surgery resection-induced hyperinflammatory microenvironment is a key factor that leads to postsurgical cancer recurrence. Herein, we developed a dual-functional nanodrug Asp@cLANVs for postsurgical recurrence inhibition by loading the classical anti-inflammatory drug aspirin (Asp) into cross-linked lipoic acid nanovesicles (cLANVs). The Asp@cLANVs can not only kill residual cancer cells at the doses comparable to common cytotoxic drugs by synergistic interaction between Asp and cLANVs, but also improve the postsurgical inflammatory microenvironment by their strongly synergistic anti-inflammation activity between Asp and cLANVs. Using mice bearing partially removed NCI-H460 tumors, we found that Asp@cLANVs gave a much lower recurrence rate (33.3%) compared with the first-line cytotoxic drug cisplatin (100%), and no mice died for at least 60 days after Asp@cLANV treatment while no mouse survived beyond day 43 in the cisplatin group. This dual-functional nanodrug constructs the first example that combines residual cancer cell killing and postoperative inflammation microenvironment improvement to suppress postsurgical cancer recurrence.
Collapse
Affiliation(s)
- Pei Jing
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P.R. China.,Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, P.R. China
| | - Yuling Luo
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, P.R. China
| | - Yun Chen
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| | - Jiangbing Tan
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P.R. China
| | - Chunyan Liao
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P.R. China
| | - Shiyong Zhang
- College of Biomedical Engineering and National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P.R. China
| |
Collapse
|
33
|
Xie Y, Liu M, Cai C, Ye C, Guo T, Yang K, Xiao H, Tang X, Liu H. Recent progress of hydrogel-based local drug delivery systems for postoperative radiotherapy. Front Oncol 2023; 13:1027254. [PMID: 36860309 PMCID: PMC9969147 DOI: 10.3389/fonc.2023.1027254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/02/2023] [Indexed: 02/15/2023] Open
Abstract
Surgical resection and postoperative radiotherapy remained the most common therapeutic modalities for malignant tumors. However, tumor recurrence after receiving such combination is difficult to be avoided because of high invasiveness and radiation resistance of cancer cells during long-term therapy. Hydrogels, as novel local drug delivery systems, presented excellent biocompatibility, high drug loading capacity and sustained drug release property. Compared with conventional drug formulations, hydrogels are able to be administered intraoperatively and directly release the entrapped therapeutic agents to the unresectable tumor sites. Therefore, hydrogel-based local drug delivery systems have their unique advantages especially in sensitizing postoperative radiotherapy. In this context, classification and biological properties of hydrogels were firstly introduced. Then, recent progress and application of hydrogels for postoperative radiotherapy were summarized. Finally, the prospects and challenges of hydrogels in postoperative radiotherapy were discussed.
Collapse
Affiliation(s)
- Yandong Xie
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China,Department of Neurosurgery, The Suqian Clinical College of Xuzhou Medical University, Suqian, China
| | - Mingxi Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Chang Cai
- Department of Neurosurgery, The Suqian Clinical College of Xuzhou Medical University, Suqian, China
| | - Chengkun Ye
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tangjun Guo
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kun Yang
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China,*Correspondence: Hongyi Liu, ; Xianglong Tang, ; Hong Xiao,
| | - Xianglong Tang
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China,Department of Neuro-Psychiatric Institute, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China,*Correspondence: Hongyi Liu, ; Xianglong Tang, ; Hong Xiao,
| | - Hongyi Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China,*Correspondence: Hongyi Liu, ; Xianglong Tang, ; Hong Xiao,
| |
Collapse
|
34
|
Dual drug-loaded hydrogels with pH-responsive and antibacterial activity for skin wound dressing. Colloids Surf B Biointerfaces 2023; 222:113063. [PMID: 36502601 DOI: 10.1016/j.colsurfb.2022.113063] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Antibacterial and hemostatic properties are essential for wound healing dressing. In this study, a new type of hydrogel composed of gelatin methacryloyl (GelMA) and hyaluronic acid-aldehyde (HA-CHO) is fabricated by photo-crosslinking and respectively loaded with a single drug gentamicin sulfate (GS), and two drugs of GS and lysozyme (LZM). The composite hydrogel of GelMA and HA-CHO is successfully synthesized by the aldehyde and Schiff base reactions. The structures and compositions of the hydrogels with and without drug loaded are characterized by FT-IR, 1H NMR, and XPS. Furthermore, the microstructure and swelling behaviour of hydrogels prove that the content of HA-CHO has a significant role in the formation of hydrogels with dense porous structures and super absorbent. pH 7.4 and pH 5.0 conditions are used to evaluate the drug release behaviour of the obtained hydrogels. The released amount of GS of the drug-loaded hydrogels in the acidic buffer is more than that of the physiological environment because of the cleaved Schiff base bonds and the electrostatic interaction. Especially for the dual drug-loaded hydrogel GelMA/HA-CHO/GS/LZM, the released ratio of GS is elevated from 59 % in pH 7.4 buffer to about 78 % in pH 5.0 buffer within the first 6 h, which verifies the excellent pH-stimulus responsiveness. These endow the GS-LZM dual drug-loaded hydrogels with superior antibacterial efficiencies to that of the single GS drug-loaded hydrogels, no drug-loaded hydrogels, and SEBS control, especially in inhibiting S. aureus in a lower concentration of 106 CFU mL-1, which can be attributed to the synergistic effect of LZM and GS. For S. aureus at 106 CFU mL-1, the bacterial survival of GelMA/HA-CHO/GS/LZM is 1.1 %, which shows outstanding antibacterial effect. Hence, the drug-loaded hydrogels, especially the dual drug-loaded hydrogels with pH-responsive, antibacterial, and hemostatic properties have great potential as wound healing materials.
Collapse
|
35
|
Lu X, Li X, Yu J, Ding B. Nanofibrous hemostatic materials: Structural design, fabrication methods, and hemostatic mechanisms. Acta Biomater 2022; 154:49-62. [PMID: 36265792 DOI: 10.1016/j.actbio.2022.10.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/16/2022] [Accepted: 10/12/2022] [Indexed: 12/14/2022]
Abstract
Development of rapid and effective hemostatic materials has always been the focus of research in the healthcare field. Nanofibrous materials which recapitulate the delicate nano-topography feature of fibrin fibers produced during natural hemostatic process, offer large length-to-diameter ratio and surface area, tunable porous structure, and precise control in architecture, showing great potential for staunching bleeding. Here we present a comprehensive review of advances in nanofibrous hemostatic materials, focusing on the following three important parts: structural design, fabrication methods, and hemostatic mechanisms. This review begins with an introduction to the physiological hemostatic mechanism and current commercial hemostatic agents. Then, it focuses on recent progress in electrospun nanofibrous hemostatic materials in terms of composition and structure control, surface modification, and in-situ deposition. The article emphasizes the development of three-dimensional (3D) electrospun nanofibrous materials and their emerging evolution for improving hemostatic function. Next, it discusses the fabrication of self-assembling peptide or protein-mimetic peptide nanofibers, co-assembling supramolecular nanofibers, as well as other nanofibrous hemostatic agents. Further, the article highlights the external and intracavitary hemostatic management based on various nanofiber aggregates. In the end, this review concludes with the current challenges and future perspectives of nanofibrous hemostatic materials. STATEMENT OF SIGNIFICANCE: This article reviews recent advances in nanofibrous hemostatic materials including fabrication methods, composition and structural control, performance improvement, and hemostatic mechanisms. A variety of methods including electrospinning, self-assembly, grinding and refining, template synthesis, and chemical vapor deposition, have been developed to prepare nanofibrous materials. These methods provide robustness in control of the nanofiber architecture in the forms of hydrogels, two-dimensional (2D) membranes, 3D sponges, or composites, showing promising potential in the external and intracavitary hemostasis and wound healing applications. This review will be of great interest to the broad readers in the field of hemostatic materials and multifunctional biomaterials.
Collapse
Affiliation(s)
- Xuyan Lu
- Innovation Center for Textile Science and Technology, College of Textiles, Donghua University, Shanghai 201620, China
| | - Xiaoran Li
- Innovation Center for Textile Science and Technology, College of Textiles, Donghua University, Shanghai 201620, China.
| | - Jianyong Yu
- Innovation Center for Textile Science and Technology, College of Textiles, Donghua University, Shanghai 201620, China
| | - Bin Ding
- Innovation Center for Textile Science and Technology, College of Textiles, Donghua University, Shanghai 201620, China.
| |
Collapse
|
36
|
Zhang Q, Wang X, Kuang G, Yu Y, Zhao Y. Photopolymerized 3D Printing Scaffolds with Pt(IV) Prodrug Initiator for Postsurgical Tumor Treatment. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9784510. [PMID: 36111316 PMCID: PMC9448443 DOI: 10.34133/2022/9784510] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/01/2022] [Indexed: 12/20/2022]
Abstract
Biomedical scaffolds have shown great success in postsurgical tumor treatment; their current efforts are focusing on eradicating residual tumor cells and circulating tumor cells and simultaneously repairing postoperative tissue defects. Herein, we report a novel photopolymerized 3D scaffold with Pt(IV) prodrug initiator to achieve the desired features for tumor comprehensive therapy. The Pt-GelMA scaffold was fabricated from the microfluidic 3D printing of methacrylate gelatin (GelMA) bioinks through a Pt(IV)-induced photocrosslinked process without any other additional photoinitiator and chemotherapeutic drug. Thus, the resultant scaffold displayed efficient cell killing ability against breast cancer cells in vitro and significantly inhibited the local tumor growth and distant metastases on an orthotopic postoperative breast cancer model in vivo. Besides, benefiting from their ordered porous structures and favorable biocompatibility, the scaffolds supported the cell attachment, spreading, and proliferation of normal cells in vitro; could facilitate the nutrient transportation; and induced new tissue ingrowth for repairing tissue defects caused by surgery. These properties indicate that such 3D printing scaffold is a promising candidate for efficient postoperative tumor treatment in the practical application.
Collapse
Affiliation(s)
- Qingfei Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Xiaocheng Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Gaizhen Kuang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yunru Yu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| |
Collapse
|
37
|
Bao G, Gao Q, Cau M, Ali-Mohamad N, Strong M, Jiang S, Yang Z, Valiei A, Ma Z, Amabili M, Gao ZH, Mongeau L, Kastrup C, Li J. Liquid-infused microstructured bioadhesives halt non-compressible hemorrhage. Nat Commun 2022; 13:5035. [PMID: 36028516 PMCID: PMC9418157 DOI: 10.1038/s41467-022-32803-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/17/2022] [Indexed: 11/30/2022] Open
Abstract
Non-compressible hemorrhage is an unmet clinical challenge that accounts for high mortality in trauma. Rapid pressurized blood flows under hemorrhage impair the function and integrity of hemostatic agents and the adhesion of bioadhesive sealants. Here, we report the design and performance of bioinspired microstructured bioadhesives, formed with a macroporous tough xerogel infused with functional liquids. The xerogel can rapidly absorb interfacial fluids such as whole blood and promote blood clotting, while the infused liquids facilitate interfacial bonding, sealing, and antibacterial function. Their synergy enables the bioadhesives to form tough adhesion on ex vivo human and porcine tissues and diverse engineered surfaces without the need for compression, as well as on-demand instant removal and storage stability. We demonstrate a significantly improved hemostatic efficacy and biocompatibility in rats and pigs compared to non-structured counterparts and commercial products. This work opens new avenues for the development of bioadhesives and hemostatic sealants.
Collapse
Affiliation(s)
- Guangyu Bao
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Qiman Gao
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Massimo Cau
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Nabil Ali-Mohamad
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Mitchell Strong
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Shuaibing Jiang
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Zhen Yang
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Amin Valiei
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
| | - Zhenwei Ma
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Marco Amabili
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Zu-Hua Gao
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Luc Mongeau
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Christian Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.
- Blood Research Institute, Versiti, Milwaukee, WI, USA.
- Department of Surgery, Division of Trauma and Acute Care Surgery, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Jianyu Li
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada.
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada.
- Department of Surgery, McGill University, Montreal, QC, Canada.
| |
Collapse
|
38
|
Chen S, Song Y, Yan X, Dong L, Xu Y, Xuan S, Shu Q, Cao B, Hu J, Xing H, Wu W, Zha Z, Lu Y. Injectable magnetic montmorillonite colloidal gel for the postoperative treatment of hepatocellular carcinoma. J Nanobiotechnology 2022; 20:381. [PMID: 35986283 PMCID: PMC9392261 DOI: 10.1186/s12951-022-01559-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
Bioactive materials have been extensively developed for the adjuvant therapy of cancer. However, few materials can meet the requirements for the postoperative resection of hepatocellular carcinoma (HCC) due to massive bleeding and high recurrence. In particular, combination therapy for HCC has been highly recommended in clinical practice, including surgical resection, interventional therapy, ablation therapy and chemotherapy. Herein, an injectable magnetic colloidal gel (MCG) was developed by controllable electrostatic attraction between clinically available magnetic montmorillonites and amphoteric gelatin nanoparticles. The optimized MCG exhibited an effective magnetic heating effect, remarkable rheological properties, and high gel network stability, realizing the synergistic treatment of postoperative HCC by stimuli-responsive drug delivery, hemostasis and magnetic hyperthermia. Furthermore, a minimal invasive MCG-induced interventional magnetic hyperthermia therapy (MHT) under ultrasound guidance was realized on hepatic tumor rabbits, providing an alternative therapeutics to treat the postoperative recurrence. Overall, MCG is a clinically available injectable formulation for adjuvant therapy after HCC surgical resection.
Collapse
|
39
|
Hou Y, Fei Y, Liu Z, Liu Y, Li M, Luo Z. Black phosphorous nanomaterials as a new paradigm for postoperative tumor treatment regimens. J Nanobiotechnology 2022; 20:366. [PMID: 35953821 PMCID: PMC9367102 DOI: 10.1186/s12951-022-01579-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/24/2022] [Indexed: 12/02/2022] Open
Abstract
Surgery is currently a mainstream treatment modality for various solid tumor indications. However, aggressive resection of tumor tissues frequently causes postoperative complications, which severely undermine the well-being of patients. Moreover, the residue tumor cells may substantially increase the risk of local and distant tumor relapse. The recent development in black phosphorus (BP)-based nanomaterials offers a promising opportunity to address these clinical challenges. BP is an emerging nanomaterial with excellent biocompatibility and versatile functionality, which has already demonstrated great potential for a variety of biomedical applications including tumor therapy and tissue engineering. In this review, the recent advances in BP-based nanobiomaterials for the post-surgery treatment of solid tumor have been summarized, while specific emphasis was placed on their capability to continuously inhibit residue tumor growth at the surgery site as well as stimulating various healing mechanisms, aiming to preventing tumor relapse while promoting the healing of surgery-induced traumatic soft/hard tissue injuries. It is anticipated that the nanoengineered BP-based materials may open new avenues to tackle those clinical challenges in surgical treatment of solid tumors.
Collapse
Affiliation(s)
- Yanhua Hou
- Chongqing Engineering Research Center of Pharmaceutical Science, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Zehong Liu
- Chongqing Engineering Research Center of Pharmaceutical Science, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, China
| | - Yingqi Liu
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China. .,111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
40
|
Gong Y, Chen W, Chen X, He Y, Jiang H, Zhang X, Pan L, Ni B, Yang F, Xu Y, Zhang Q, Zhou L, Cheng Y. An Injectable Epigenetic Autophagic Modulatory Hydrogel for Boosting Umbilical Cord Blood NK Cell Therapy Prevents Postsurgical Relapse of Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201271. [PMID: 35712750 PMCID: PMC9376812 DOI: 10.1002/advs.202201271] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/03/2022] [Indexed: 06/15/2023]
Abstract
Triple-negative breast cancer (TNBC) exhibits resistance to conventional treatments due to the presence of cancer stem cells (CSCs), causing postsurgical relapse and a dismal prognosis. Umbilical cord blood natural killer (UCB-NK) cell-based immunotherapy represents a promising strategy for cancer treatment. However, its therapeutic efficacy is greatly restrained by downregulation of the NK cell activation ligand MHC class I-related chain A/B (MICA/B) and autophagy-mediated degradation of NK cell-derived granzyme B (GZMB) in CSCs. Herein, it is demonstrated that suberoylanilide hydroxamic acid (SAHA) epigenetically downregulates let-7e-5p and miR-615-3p to increase MICA/B expression and that 3-methyl adenine (3MA) inhibits autophagy-mediated GZMB degradation, thereby sensitizing breast CSCs to UCB-NK cells. Then, an injectable hydrogel is designed to codeliver SAHA and 3MA to enhance UCB-NK cell infusion efficacy in TNBC. The hydrogel precursors can be smoothly injected into the tumor resection bed and form a stable gel in situ, allowing for a pH-sensitive sustained release of SAHA and 3MA. Moreover, UCB-NK cell infusion in combination with the hydrogel efficiently controls postsurgical relapse of TNBC. In addition, the hydrogel exhibits good hemostasis and wound-healing functions. Therefore, the work provides proof of concept that an injectable epigenetic autophagic modulatory hydrogel augments UCB-NK cell therapy to combat postsurgical relapse of TNBC.
Collapse
Affiliation(s)
- Yihang Gong
- Department of Hepatic Surgery and Liver Transplantation Center & Guangdong Provincial Key Laboratory of Liver Disease ResearchThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Wenjie Chen
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Xiuxing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Medical OncologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Yizhan He
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Hua Jiang
- Department of Breast & Thyroid SurgeryThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Xijian Zhang
- Department of Hepatic Surgery and Liver Transplantation Center & Guangdong Provincial Key Laboratory of Liver Disease ResearchThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Lijie Pan
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Beibei Ni
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Fan Yang
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Yan Xu
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Qi Zhang
- Biotherapy Centre & Cell‐Gene Therapy Translational Medicine Research CentreThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Lei Zhou
- Guangzhou Key Laboratory of Spine Disease Prevention and TreatmentDepartment of Spine SurgeryThe Third Affiliated HospitalGuangzhou Medical UniversityGuangzhou510150China
| | - Yusheng Cheng
- Department of Hepatic Surgery and Liver Transplantation Center & Guangdong Provincial Key Laboratory of Liver Disease ResearchThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| |
Collapse
|
41
|
Electrospun Silk Fibroin/Polylactic-co-glycolic Acid/Black Phosphorus Nanosheets Nanofibrous Membrane with Photothermal Therapy Potential for Cancer. Molecules 2022; 27:molecules27144563. [PMID: 35889436 PMCID: PMC9317578 DOI: 10.3390/molecules27144563] [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: 05/19/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 01/27/2023] Open
Abstract
Photothermal therapy is a promising treating method for cancers since it is safe and easily controllable. Black phosphorus (BP) nanosheets have drawn tremendous attention as a novel biodegradable thermotherapy material, owing to their excellent biocompatibility and photothermal properties. In this study, silk fibroin (SF) was used to exfoliate BP with long-term stability and good solution-processability. Then, the prepared BP@SF was introduced into fibrous membranes by electrospinning, together with SF and polylactic-co-glycolic acid (PLGA). The SF/PLGA/BP@SF membranes had relatively smooth and even fibers and the maximum stress was 2.92 MPa. Most importantly, the SF/PLGA/BP@SF membranes exhibited excellent photothermal properties, which could be controlled by the BP@SF content and near infrared (NIR) light power. The temperature of SF/PLGA/BP@SF composite membrane was increased by 15.26 °C under NIR (808 nm, 2.5 W/cm2) irradiation for 10 min. The photothermal property of SF/PLGA/BP@SF membranes significantly killed the HepG2 cancer cells in vitro, indicating its good potential for application in local treatment of cancer.
Collapse
|
42
|
Bhattacharjee B, Mukherjee R, Haldar J. Biocompatible Hemostatic Sponge Exhibiting Broad-Spectrum Antibacterial Activity. ACS Biomater Sci Eng 2022; 8:3596-3607. [PMID: 35802178 DOI: 10.1021/acsbiomaterials.2c00410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Hemorrhage during accidents or surgery is a significant challenge that can contribute to mortality. This is further aggravated due to bacterial infections at the injured site. Therefore, rapid application of a hemostatic and antibacterial material is highly necessary as a pretreatment for patients' survival. Herein, we have developed a hemostatic sponge (Hemobac) through amide crosslinking of gelatin and an N-(2-hydroxy) propyl-3-trimethylammonium chitosan (HTCC)-silver chloride nanocomposite (QAm1-Ag0.1) to mitigate bacterial infections, while aiding hemostasis. This Hemobac sponge completely eradicated (∼4-5 log) a wide range of Gram-positive and Gram-negative bacteria encompassing various clinical isolates within 6 h. The antihemorrhagic ability of Hemobac was ascertained through SEM images, which exhibited the presence of agglomerated blood cells onto the sponge with a significantly low blood-clotting index value (∼23 ± 1). Notably, Hemobac reduced the blood loss by ∼70-80% in the liver puncture model and femoral vein injury model in mice, displaying its improved hemostatic ability over a marketed gelatin-based sponge. Negligible hemolytic activity (∼6%) and retained healthy morphology of mammalian cells were observed upon exposure to the Hemobac sponge. Minimal immune response was noticed at the Hemobac-treated wound in mice through histopathology analysis. Collectively, these findings indicate that this biocompatible Hemobac sponge can stop the bleeding instantaneously and combat bacterial infections.
Collapse
Affiliation(s)
- Brinta Bhattacharjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Riya Mukherjee
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India.,School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
43
|
Fang Y, Liu Z, Wang H, Luo X, Xu Y, Chan HF, Lv S, Tao Y, Li M. Implantable Sandwich-like Scaffold/Fiber Composite Spatiotemporally Releasing Combretastatin A4 and Doxorubicin for Efficient Inhibition of Postoperative Tumor Recurrence. ACS APPLIED MATERIALS & INTERFACES 2022; 14:27525-27537. [PMID: 35687834 DOI: 10.1021/acsami.2c02103] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Tumor recurrence is a critical conundrum in the postoperative therapy, on account of severe bleeding with disseminated tumor cells, residual tumor cells, and the rich nutrient and oxygen supply transported to tumors by the abundant blood vessels. Biodegradable drug-loaded implants, inserted in the resection cavity right away upon the surgery, possess bleeding prevention and efficient chemotherapeutic capabilities, considered to be a promising strategy to efficiently inhibit the recurrence of the solid tumor. Here, we developed a sandwich-like composite consisting of the combretastatin A4 (CA4)-loaded 3D-printed scaffold and doxorubicin (DOX)-loaded electrospun fiber (Scaffold-CA4@Fiber-DOX), presenting hemostatic, chemotherapeutic, and antibacterial potencies. The lyophilized 3D-printed scaffold with a porous structure rapidly absorbed and clotted the blood cells and disseminated tumor cells to prevent bleeding and tumor metastasis. Subsequently, the preferentially released CA4 from the scaffold disrupted the microtubules of the vascular endothelial cell, resulting in vascular deformation and consequent insufficient nutrient supply to the solid tumor. The sustained release of DOX from the sandwiched electrospun fiber dramatically inhibited the peripheral tumor cell proliferation. This all-in-one multifunctional implant system, combining efficient vascular disruption and chemotherapy, provides a promising strategy for postoperative tumor therapy.
Collapse
Affiliation(s)
- Youqiang Fang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zheng Liu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xing Luo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease, Guangzhou 510630, China
| |
Collapse
|
44
|
Dang W, Chen WC, Ju E, Xu Y, Li K, Wang H, Wang K, Lv S, Shao D, Tao Y, Li M. 3D printed hydrogel scaffolds combining glutathione depletion-induced ferroptosis and photothermia-augmented chemodynamic therapy for efficiently inhibiting postoperative tumor recurrence. J Nanobiotechnology 2022; 20:266. [PMID: 35672826 PMCID: PMC9171966 DOI: 10.1186/s12951-022-01454-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/05/2022] [Indexed: 12/24/2022] Open
Abstract
AbstractSurgical resection to achieve tumor-free margins represents a difficult clinical scenario for patients with hepatocellular carcinoma. While post-surgical treatments such as chemotherapy and radiotherapy can decrease the risk of cancer recurrence and metastasis, growing concerns about the complications and side effects have promoted the development of implantable systems for locoregional treatment. Herein, 3D printed hydrogel scaffolds (designed as Gel-SA-CuO) were developed by incorporating one agent with multifunctional performance into implantable devices to simplify the fabrication process for efficiently inhibiting postoperative tumor recurrence. CuO nanoparticles can be effectively controlled and sustained released during the biodegradation of hydrogel scaffolds. Notably, the released CuO nanoparticles not only function as the reservoir for releasing Cu2+ to produce intracellular reactive oxygen species (ROS) but also serve as photothermal agent to generate heat. Remarkably, the heat generated by photothermal conversion of CuO nanoparticles further promotes the efficiency of Fenton-like reaction. Additionally, ferroptosis can be induced through Cu2+-mediated GSH depletion via the inactivation of GPX4. By implanting hydrogel scaffolds in the resection site, efficient inhibition of tumor recurrence after primary resection can be achieved in vivo. Therefore, this study may pave the way for the development of advanced multifunctional implantable platform for eliminating postoperative relapsable cancers.
Graphical Abstract
Collapse
|
45
|
Ren K, He J, Qiu Y, Xu Z, Wang X, Li J, Zang S, Yang Y, Li J, Long Y, Zhang Z, Li M, He Q. A neutrophil-mediated carrier regulates tumor stemness by inhibiting autophagy to prevent postoperative triple-negative breast cancer recurrence and metastasis. Acta Biomater 2022; 145:185-199. [PMID: 35447368 DOI: 10.1016/j.actbio.2022.04.017] [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: 12/28/2021] [Revised: 03/14/2022] [Accepted: 04/09/2022] [Indexed: 02/05/2023]
Abstract
Recurrence and metastasis after resection are still the main challenges in clinical treatment of breast cancer. Residual tumor and cancer stem-like cells are the primary culprits of recurrence and metastasis. Recent research studies indicate that autophagy is a cytoprotective mechanism of tumors, which maintains the stemness of cancer cells and promotes tumor proliferation and metastasis. Here, we constructed a "Trojan horse" using neutrophils as the carrier (PH-RL@NEs) to prevent the recurrence and metastasis of postoperative breast cancer. Neutrophils, as a "Trojan horse," can quickly respond to postoperative inflammation and accurately deliver drugs to the residual tumor site. The inflammation-triggered "Trojan horse" was then opened to release the liposomes containing the chemotherapeutic drug paclitaxel (PTX) and the autophagy inhibitor hydroxychloroquine (HCQ). We found that HCQ could effectively inhibit tumor cell autophagy, interfere with tumor epithelial-mesenchymal transition, and reduce the tumor stem cell-like population. In the orthotopic 4T1 postoperative recurrence models, PTX and HCQ synergistically killed tumors and regulated the stemness of tumor cells, thereby significantly inhibiting tumor recurrence and metastasis. Our work proved that the inhibition of autophagy to reduce tumor stemness is feasible and effective, which opens up a new prospect for postoperative tumor treatment. STATEMENT OF SIGNIFICANCE: The present study aimed to solve the issues of postoperative recurrence and metastasis of breast cancer and low efficiency of drug administration after surgery. For this purpose, we constructed neutrophils containing hydroxychloroquine (HCQ) and paclitaxel (PTX) co-loaded liposomes (PH-RL@NEs), which for the first time regulated the stemness of tumor cells by inhibiting autophagy, thereby inhibiting postoperative recurrence and metastasis of breast cancer cells. The results showed that PH-RL@NEs enhanced the targeted drug delivery efficiency, with the help of postoperative inflammation chemotaxis of neutrophils. HCQ effectively inhibited autophagy of tumor cells and reduced tumor stem cell-like cells, thus improving the therapeutic effect in the 4T1 in situ postoperative recurrence model.
Collapse
Affiliation(s)
- Kebai Ren
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiao He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yue Qiu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhuping Xu
- West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xuhui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiaxin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shuya Zang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yiliang Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiaxin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yang Long
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Med-X Center for Materials, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
46
|
Chen Y, Hao Y, Mensah A, Lv P, Wei Q. Bio-inspired hydrogels with fibrous structure: A review on design and biomedical applications. BIOMATERIALS ADVANCES 2022; 136:212799. [PMID: 35929334 DOI: 10.1016/j.bioadv.2022.212799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Numerous tissues in the human body have fibrous structures, including the extracellular matrix, muscles, and heart, which perform critical biological functions and have exceptional mechanical strength. Due to their high-water content, softness, biocompatibility and elastic nature, hydrogels resemble biological tissues. Traditional hydrogels, on the other hand, have weak mechanical properties and lack tissue-like fibrous structures, limiting their potential applications. Thus, bio-inspired hydrogels with fibrous architectures have piqued the curiosity of biomedical researchers. Here, we review fabrication strategies for fibrous hydrogels and their recent progress in the biomedical fields of wound dressings, drug delivery, tissue engineering scaffolds and bioadhesives. Challenges and future perspectives are also discussed.
Collapse
Affiliation(s)
- Yajun Chen
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China
| | - Yi Hao
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China
| | - Alfred Mensah
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China
| | - Pengfei Lv
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China
| | - Qufu Wei
- Key Laboratory of Eco-textiles, Ministry of Education, Jiangnan University, Wuxi, People's Republic of China.
| |
Collapse
|
47
|
Kefayat A, Hosseini M, Ghahremani F, Jolfaie NA, Rafienia M. Biodegradable and biocompatible subcutaneous implants consisted of pH-sensitive mebendazole-loaded/folic acid-targeted chitosan nanoparticles for murine triple-negative breast cancer treatment. J Nanobiotechnology 2022; 20:169. [PMID: 35361226 PMCID: PMC8973744 DOI: 10.1186/s12951-022-01380-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/17/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Mebendazole (MBZ) is a well-known anti-parasite drug with significant anti-cancer properties. However, MBZ exhibits low solubility, limited absorption efficacy, extensive first-pass effect, and low bioavailability. Therefore, multiple oral administration of high dose MBZ is required daily for achieving the therapeutic serum level which can cause severe side effects and patients' non-compliance. METHOD In the present study, MBZ-loaded/folic acid-targeted chitosan nanoparticles (CS-FA-MBZ) were synthesized, characterized, and used to form cylindrical subcutaneous implants for 4T1 triple-negative breast tumor (TNBC) treatment in BALB/c mice. The therapeutic efficacy of the CS-FA-MBZ implants was investigated after subcutaneous implantation in comparison with Control, MBZ (40 mg/kg, oral administration, twice a week for 2 weeks), and CS-FA implants, according to 4T1 tumors' growth progression, metastasis, and tumor-bearing mice survival time. Also, their biocompatibility was evaluated by blood biochemical analyzes and histopathological investigation of vital organs. RESULTS The CS-FA-MBZ implants were completely degraded 15 days after implantation and caused about 73.3%, 49.2%, 57.4% decrease in the mean tumors' volume in comparison with the Control (1050.5 ± 120.7 mm3), MBZ (552.4 ± 76.1 mm3), and CS-FA (658.3 ± 88.1 mm3) groups, respectively. Average liver metastatic colonies' number per microscope field at the CS-FA-MBZ group (2.3 ± 0.7) was significantly (P < 0.05) lower than the Control (9.6 ± 1.7), MBZ (5.0 ± 1.5), and CS-FA (5.2 ± 1) groups. In addition, the CS-FA-MBZ treated mice exhibited about 52.1%, 27.3%, and 17% more survival days after the cancer cells injection in comparison with the Control, MBZ, and CS-FA groups, respectively. Moreover, the CS-FA-MBZ implants were completely biocompatible based on histopathology and blood biochemical analyzes. CONCLUSION Taking together, CS-FA-MBZ implants were completely biodegradable and biocompatible with high therapeutic efficacy in a murine TNBC model.
Collapse
Affiliation(s)
- Amirhosein Kefayat
- Cancer Prevention Research Center, Department of Oncology, Isfahan University of Medical Sciences, 81746-73461, Isfahan, Iran.,Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Hosseini
- Department of Chemistry, Amirkabir University of Technology (Tehran Polytechnic), 1591634311, Tehran, Iran
| | - Fatemeh Ghahremani
- Department of Medical Physics and Radiotherapy, School of Paramedicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafise Arbab Jolfaie
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Rafienia
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
48
|
Yan X, Sun T, Song Y, Peng W, Xu Y, Luo G, Li M, Chen S, Fang WW, Dong L, Xuan S, He T, Cao B, Lu Y. In situ Thermal-Responsive Magnetic Hydrogel for Multidisciplinary Therapy of Hepatocellular Carcinoma. NANO LETTERS 2022; 22:2251-2260. [PMID: 35254836 DOI: 10.1021/acs.nanolett.1c04413] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Current surgical single modality treatments for hepatocellular carcinoma (HCC) were restricted by recurrence, blood loss, significant trauma, and poor prognostic. Although multidisciplinary strategies for HCC treatment have been highly recommended by the clinical guidelines, there was limited choice of materials and treatments. Herein, we reported an in situ formed magnetic hydrogel with promising bioapplicable thermal-responsiveness, strong adhesion in wet conditions, high magnetic hyperthermia, and biocompatibility, leading to efficient HCC multidisciplinary treatment including postoperative treatment and transarterial embolization therapy. In vivo results indicated that this hydrogel could reduce the postoperative recurrence rate. The hemostatic ability of the thermal-responsive hydrogel was further demonstrated in both the liver scratch model and liver tumor resection. Computed tomography imaging suggested that the hydrogel could completely embolize the arterial vessels of rabbit liver tumor by vascular intervention operation, which could serve as multidisciplinary responsive materials to external magnetic field and body temperature for HCC treatment.
Collapse
Affiliation(s)
- Xu Yan
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Tianci Sun
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Yonghong Song
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Wei Peng
- Department of General Surgery, Department of Interventional Radiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, China
| | - Yunjun Xu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Guangyi Luo
- Department of General Surgery, Department of Interventional Radiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, China
| | - Min Li
- Department of General Surgery, Department of Interventional Radiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, China
| | - Sheng Chen
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Wei-Wei Fang
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Liang Dong
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Shouhu Xuan
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Tao He
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Baoqiang Cao
- Department of General Surgery, Department of Interventional Radiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, China
| | - Yang Lu
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| |
Collapse
|
49
|
Wang N, Lin JY, Luo SH, Zhou YJ, Yang K, Chen RH, Yang GX, Wang ZY. Furanonyl amino acid derivatives as hemostatic drugs: design, synthesis and hemostasis performance. Amino Acids 2022; 54:989-999. [PMID: 35305164 DOI: 10.1007/s00726-022-03155-3] [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: 09/26/2021] [Accepted: 03/07/2022] [Indexed: 11/01/2022]
Abstract
Using 3,4-dihalo-2(5H)-furanones and easily available hemostatic drugs, such as tranexamic acid (TA), 4-aminomethylbenzoic acid (ABA), aminocaproic acid (AA) as starting materials, serial multi-functional molecules 2(5H)-furanonyl amino acids are designed by the combination of different pharmacophores, and successfully synthesized by a transition metal-free Michael addition-elimination reaction. The reaction is carried out under mild conditions with ethanol-dichloromethane as solvent and only stirring at room temperature for 24 h, and the yield can be up to 91%. All products are well characterized by infrared spectroscopy (IR), nuclear magnetic resonance (NMR), high-resolution mass spectra (HRMS). Ten typical target compounds among them are selected out for the experiments of hemostasis performance by the evaluation of in vitro clot formation model and liver hemorrhage model. The test results show that, their hemostasis effect is better than the original drugs. Especially the target compound G, a TA derivative from 5-borneoloxy-3,4-dibromo-2(5H)-furanone, has the best hemostasis effect among all the tested compounds. These obtained target molecules are expected to be used as multi-functional hemostatic drugs.
Collapse
Affiliation(s)
- Neng Wang
- School of Chemistry, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, South China Normal University, Guangzhou, 510006, People's Republic of China.,School of Chemistry, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou, 510006, People's Republic of China.,School of Chemistry, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, South China Normal University, Guangzhou, 510006, People's Republic of China
| | - Jian-Yun Lin
- School of Chemistry, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, South China Normal University, Guangzhou, 510006, People's Republic of China.,School of Chemistry, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou, 510006, People's Republic of China.,School of Chemistry, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, South China Normal University, Guangzhou, 510006, People's Republic of China
| | - Shi-He Luo
- School of Chemistry, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, South China Normal University, Guangzhou, 510006, People's Republic of China. .,Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, 381 Wushan Road, Guangzhou, 510640, People's Republic of China. .,School of Chemistry, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou, 510006, People's Republic of China. .,School of Chemistry, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, South China Normal University, Guangzhou, 510006, People's Republic of China.
| | - Yong-Jun Zhou
- School of Chemistry, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, South China Normal University, Guangzhou, 510006, People's Republic of China.,School of Chemistry, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou, 510006, People's Republic of China.,School of Chemistry, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, South China Normal University, Guangzhou, 510006, People's Republic of China
| | - Kai Yang
- School of Chemistry, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, South China Normal University, Guangzhou, 510006, People's Republic of China. .,College of Pharmacy, Gannan Medical University, Ganzhou, 341000, People's Republic of China. .,School of Chemistry, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou, 510006, People's Republic of China. .,School of Chemistry, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, South China Normal University, Guangzhou, 510006, People's Republic of China.
| | - Ren-Hong Chen
- Guangdong Food and Drug Vocational College, Guangzhou, 510520, People's Republic of China.
| | - Guo-Xian Yang
- School of Chemistry, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, South China Normal University, Guangzhou, 510006, People's Republic of China.,School of Chemistry, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou, 510006, People's Republic of China.,School of Chemistry, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, South China Normal University, Guangzhou, 510006, People's Republic of China
| | - Zhao-Yang Wang
- School of Chemistry, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, South China Normal University, Guangzhou, 510006, People's Republic of China. .,Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, 381 Wushan Road, Guangzhou, 510640, People's Republic of China. .,School of Chemistry, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou, 510006, People's Republic of China. .,School of Chemistry, GDMPA Key Laboratory for Process Control and Quality Evaluation of Chiral Pharmaceuticals, South China Normal University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
50
|
Wang Y, Niu W, Qu X, Lei B. Bioactive Anti-Inflammatory Thermocatalytic Nanometal-Polyphenol Polypeptide Scaffolds for MRSA-Infection/Tumor Postsurgical Tissue Repair. ACS APPLIED MATERIALS & INTERFACES 2022; 14:4946-4958. [PMID: 35073045 DOI: 10.1021/acsami.1c21082] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Postsurgical tumor recurrence, infection, and tissue defect are still the challenges in clinical medicine. The development of multifunctional biomaterial scaffolds with a microenvironment-responsive tumor-infection therapy-tissue repair is highly desirable. Herein, we report a bioactive, injectable, adhesive, self-healing, antibacterial, and anti-inflammatory metal-polyphenol polypeptide nanocomposite scaffold (PEAPF) with temporal-spatial-controlled inflammation-triggered therapeutic properties for efficient infection and postsurgical tumor therapy and skin repair. PEAPF scaffolds showed sustained and inherent inflammation-triggered Fenton catalysis and mild thermochemical effect for specifically inhibiting tumor recurrence in vitro and in vivo. The PEAPF scaffolds significantly facilitated skin tissue regeneration in MRSA-infected chronic wounds and postsurgical tissue defects after tumor resection. This study presents the multifunctional scaffold-based safe and efficient therapeutic strategy to prevent local tumor recurrence and enhance postsurgical tissue regeneration.
Collapse
Affiliation(s)
- Yidan Wang
- Frontier Institute of Science and Technology, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China
| | - Wen Niu
- Frontier Institute of Science and Technology, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China
| | - Xiaoyan Qu
- Frontier Institute of Science and Technology, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China
| | - Bo Lei
- Frontier Institute of Science and Technology, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710054, China
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710054, China
| |
Collapse
|