1
|
Zhou Y, Liu M, He C, Lin J, Chen Y, Yu M, Jiang Y, Peng X. Innovative liquid embolic agents based on deep eutectic solvent: Rapid gelation in situ via solvent exchange with water for endovascular embolization. Bioact Mater 2025; 48:550-563. [PMID: 40104022 PMCID: PMC11914998 DOI: 10.1016/j.bioactmat.2025.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/04/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Current liquid embolic agents face several challenges, including poor biocompatibility and vascular recanalization. Herein, we propose an innovative liquid embolic agent composed of a coenzyme-based polymer (poly lipoic acid, PLA) and a biocompatible solvent (deep eutectic solvent, DES). The agent undergoes phase transformation to form a stable hydrogel in situ through solvent exchange with water, thereby enabling safe and effective embolization. First, DES is obtained by heating a mixture of choline chloride (ChCl) and glycerol (Gly). Subsequently, lipoic acid (LA) is incorporated into the DES and heated to produce the PLA/DES complex. Owing to the strong hydrogen bonding between the DES and PLA, the DES acts as a solvent while also inhibiting PLA depolymerization. Upon contact with blood, most of the DES exchange with water, whereas some amount of ChCl integrates within the PLA via strong hydrogen bonding. This hydrogen bonding not only prevents PLA depolymerization but also reinforces the PLA network, resulting in a stable PLA hydrogel rather than depolymerized LA monomers. Furthermore, liquid-metal (LM) nanoparticles are incorporated to fabricate radiopaque PLA/LM/DES. PLA/LM/DES shows better in vitro hemocompatibility and cytocompatibility, milder inflammatory response in a rat model, and more effective and safer embolization in a rabbit model than a commercial embolic agent (Onyx). Thus, this work provides an innovative liquid embolic agent and broadens the biomedical applications of DES.
Collapse
Affiliation(s)
- Yitong Zhou
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
- Department of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
| | - Menghui Liu
- Department of Radiology, Hunan Engineering Research Center for Intelligent Medical Imaging of Central South University, Furong Laboratory of Central South University, Molecular Imaging Research Center of Central South University, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Chuandong He
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
| | - Jiayuan Lin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
| | - Yanlv Chen
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
| | - Mingyu Yu
- Department of Orthopedics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
| | - Yuhan Jiang
- Department of Orthopedics, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
| | - Xin Peng
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, PR China
| |
Collapse
|
2
|
Ghovvati M, Jain S, Cheng GZ, Kaneko N, Boys JA, Imahori T, De Maeseneer T, Haghniaz R, Cameron RB, Weiss AS, Annabi N. Rapid closure and hemostasis of ruptured soft tissues using a modified human tropoelastin-based sealant in preclinical models. Sci Transl Med 2025; 17:eadr6458. [PMID: 40367193 DOI: 10.1126/scitranslmed.adr6458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/16/2024] [Accepted: 04/21/2025] [Indexed: 05/16/2025]
Abstract
Treatment of injuries to soft elastic organs is often hindered by challenging anatomical features and limitations of existing sealant materials, which may lack adequate tissue adhesion, elasticity, biocompatibility, and effective hemostatic properties. To address these clinical challenges, we developed an injectable elastic sealant formulated with methacryloyl-modified human recombinant tropoelastin (MeTro) and Laponite silicate nanoplatelets (SNs). We optimized the hydrogel formulation for mechanical properties, adhesion, biocompatibility, and hemostatic properties and used visible light for cross-linking to improve safety. MeTro/SN hydrogels had increased tissue adhesion strength and burst pressure in vitro and ex vivo compared with MeTro alone or commercial sealants. The addition of SNs to the hydrogels facilitated faster blood clotting in vitro without increasing hemolysis. Applied to incisional injuries on rat lungs or aortas, MeTro/SN had burst pressures comparable to those of native tissue and greater than those of MeTro after a 7-day in vivo application. On porcine lungs, MeTro/SN also supported effective lung sealing and burst pressure similar to native lung 14 days after injury sealing. In a rodent tail hemostasis model, MeTro/SN reduced bleeding compared with MeTro. In an injured porcine lung model, early hemostasis was better than the tested commercial sealants. The results demonstrated that MeTro/SN provided effective tissue sealing and promoted hemostasis in a time frame that minimized blood loss without causing a major inflammatory response. These findings highlight the translational potential of our engineered sealant with biomimetic mechanics, durable tissue adhesion, and rapid hemostasis as a multipronged approach for the sealing and repair of traumatic injuries to soft organs.
Collapse
Affiliation(s)
- Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Saumya Jain
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - George Z Cheng
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Naoki Kaneko
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Joshua A Boys
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Taichiro Imahori
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tess De Maeseneer
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Robert B Cameron
- Division of Thoracic Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Division of Thoracic Surgery, Department of Surgery and Perioperative Care, West Los Angeles VA Medical Center, Los Angeles, CA 90095, USA
| | - Anthony S Weiss
- Charles Perkins Centre, University of Sydney, Camperdown, NSW 2006, Australia
- School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW 2006, Australia
- Sydney Nano Institute, University of Sydney, Camperdown, NSW 2006, Australia
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Kim J, Zhang Z, Albadawi H, Keum H, Mayer JL, Graf EH, Oklu R. Catheter Injectable Multifunctional Biomaterial for the Treatment of Infected Enterocutaneous Fistulas. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414642. [PMID: 39950851 PMCID: PMC12120755 DOI: 10.1002/advs.202414642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/24/2025] [Indexed: 05/31/2025]
Abstract
Enterocutaneous fistulas (ECF) are challenging to treat contributing to high morbidity and high mortality rates, significantly impacting the quality of life of the patients. Its susceptibility to antibiotic-resistant infections often leads to chronic inflammation, complicating treatment with conventional methods. Here, 18NC75-10P-1IL is reported, which is a multi-functional shear-thinning hydrogel comprised of gelatin and nanosilicates for injectability, an ionic liquid for bactericidal effects, and platelet rich fibrin fraction for pro-healing properties; this biomaterial is engineered for the treatment of ECFs. Through rigorous testing, the mechanical properties of 18NC75-10P-1IL were tailored for catheter injection to achieve durable occlusion of fistulous tracts under external pressures simulating clinical scenarios. 18NC75-10P-1IL demonstrated pro-healing effects and anti-microbial activity against highly resistant patient-derived bacteria known to be associated with ECF. Subcutaneous implantation and anorectal fistula models confirmed its biocompatibility, pro-healing, anti-inflammatory, and anti-microbial properties compared to control materials, suggesting promising potential for clinical translation in the treatment of human ECFs.
Collapse
Affiliation(s)
- Jinjoo Kim
- The Laboratory for Patient‐Inspired EngineeringMayo Clinic13400 East Shea Blvd.ScottsdaleAZ85259USA
| | - Zefu Zhang
- The Laboratory for Patient‐Inspired EngineeringMayo Clinic13400 East Shea Blvd.ScottsdaleAZ85259USA
| | - Hassan Albadawi
- The Laboratory for Patient‐Inspired EngineeringMayo Clinic13400 East Shea Blvd.ScottsdaleAZ85259USA
| | - Hyeongseop Keum
- The Laboratory for Patient‐Inspired EngineeringMayo Clinic13400 East Shea Blvd.ScottsdaleAZ85259USA
| | - Joseph L. Mayer
- The Laboratory for Patient‐Inspired EngineeringMayo Clinic13400 East Shea Blvd.ScottsdaleAZ85259USA
| | - Erin H. Graf
- Department of Laboratory Medicine and PathologyMayo Clinic5777 E Mayo BlvdPhoenixAZ85054USA
| | - Rahmi Oklu
- The Laboratory for Patient‐Inspired EngineeringMayo Clinic13400 East Shea Blvd.ScottsdaleAZ85259USA
- Division of Vascular & Interventional RadiologyMayo Clinic5777 E Mayo BlvdPhoenixAZ85054USA
| |
Collapse
|
4
|
Ahmed O, Karageorgiou J, Kumar A, Patel M, Jones J, Nezami N. Real-world clinical experience with Obsidio Conformable Embolic. CVIR Endovasc 2025; 8:36. [PMID: 40310490 PMCID: PMC12045848 DOI: 10.1186/s42155-025-00555-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Obsidio Conformable Embolic (Obsidio) is a ready-made hydrogel with unique shear-thinning properties, used for occlusion of blood flow to control bleeding or hemorrhage in the peripheral vasculature and embolization of hypervascular tumors. While pre-clinical and clinical data have demonstrated successful embolizations using Obsidio, clinical experience overall is still limited, prompting a multi-institutional field assessment survey to collect additional data on the clinical utility and procedural details from a variety of Obsidio users. The field survey collected data from 131 embolization procedures performed using Obsidio between May and November 2023 at 27 institutions within the United States. Data collection included embolization site, vessel size, any adjunctive embolics used. The primary objective of the survey was to evaluate technical success, defined as complete embolization of the target vasculature immediately following the index procedure, as confirmed by angiography. RESULTS Of the 131 embolization procedures performed, 69% (n = 90) were for hemorrhage control, 15% (n = 19) were for hypervascular tumors, and 17% (n = 22) were for other indications. Embolization of the gastroduodenal artery was the most common indication (n = 19/131; 15%). A single syringe (1 mL) or less of Obsidio was used for most cases (93%). In 33/131 cases (25%), Obsidio was combined with other embolization devices including coils (n = 25; 19%), particle-based embolics (n = 6; 4.6%), or plugs (n = 2; 1.5%). Technical success was achieved in 100% of Obsidio embolization cases (131/131 procedures). CONCLUSION Initial clinical experience demonstrated successful embolization of end-organ bleeds and hypervascular tumors utilizing Obsidio, thus making it an effective embolic agent alone or in conjunction with other embolic devices.
Collapse
Affiliation(s)
- Osman Ahmed
- Department of Radiology, University of Chicago Medical Center, Chicago, IL, USA
| | - John Karageorgiou
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Abhishek Kumar
- Department of Radiology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Mikin Patel
- Department of Radiology, University of Chicago Medical Center, Chicago, IL, USA
| | - Joshua Jones
- Boston Scientific Corporation, Marlborough, MA, USA
| | - Nariman Nezami
- Division of Vascular and Interventional Radiology, Department of Radiology, MedStar Georgetown University Hospital, 3800 Reservoir Road, NW, CCC Bldg., Room CG225, Washington, DC, 20007, USA.
- Georgetown University School of Medicine, Washington, DC, USA.
- Lombardi Comprehensive Cancer Center, Washington, DC, USA.
| |
Collapse
|
5
|
Chakraborty A, Luo W, Paul A. Nanominerals: a multifaceted biomaterial for regenerative medicine and drug delivery. Expert Opin Drug Deliv 2025:1-6. [PMID: 40211899 DOI: 10.1080/17425247.2025.2491642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/07/2025] [Indexed: 04/13/2025]
Affiliation(s)
- Aishik Chakraborty
- Department of Chemical and Biochemical Engineering, Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON, Canada
| | - Wei Luo
- School of Biomedical Engineering, The University of Western Ontario, London, ON, Canada
| | - Arghya Paul
- Department of Chemical and Biochemical Engineering, Collaborative Specialization in Musculoskeletal Health Research and Bone and Joint Institute, The University of Western Ontario, London, ON, Canada
- School of Biomedical Engineering, The University of Western Ontario, London, ON, Canada
- Department of Chemistry, The Center for Advanced Materials and Biomaterials Research, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
6
|
Peng Y, Liu H, Liang X, Cao L, Teng M, Chen H, Li Z, Peng X, Mao J, Cheng H, Liu G. Self-assembling chemodrug fiber-hydrogel for transarterial chemoembolization and radiotherapy-enhanced antitumor immunity. J Control Release 2025; 380:1-16. [PMID: 39892652 DOI: 10.1016/j.jconrel.2025.01.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
Hydrogel, as a promising embolic material for hepatocellular carcinoma (HCC), may fully embolize both major vessels and peripheral microvessels. A self-assembling hydrogel composed of chemotherapeutic drugs offers significant clinical benefits without carrier introduction. Herein, we developed a sustained drug-releasing complex hydrogel (RKT@gel), which was fabricated by the self-assembly of raltitrexed chemotherapeutic drugs (R@gel), along with the incorporation of kaempferol and tantalum nanoparticles (Ta NPs). Kaempferol enhances the mechanical strength of R@gel and inhibits hypoxia-induced angiogenesis post-embolization, improving embolization effectiveness. In addition to enabling X-ray-guided transarterial chemoembolization (TACE), Ta NPs enhance radiation sensitivity. These synergistic effects of RKT@gel not only significantly induce immunogenic cell death, thereby enhancing the activation of dendritic cells, but also activate major histocompatibility complex class I (MHC-I)-mediated antitumor immune recognition and cytotoxicity. In vivo, RKT@gel achieves enhanced tumor deposition and sustained drug release, effectively suppressing tumor progression. Additionally, when combined with radiotherapy, RKT@gel achieves efficient antitumor immunoactivation. Overall, this versatile composite hydrogel not only achieves effective embolization therapy but also substantially triggers antitumor immune responses with good biocompatibility. This multifunctional design provides a TACE-based multidisciplinary strategy for HCC.
Collapse
Affiliation(s)
- Yisheng Peng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hui Liu
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoliu Liang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Lei Cao
- Department of Pathology, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Minglei Teng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hu Chen
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhenjie Li
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuqi Peng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jingsong Mao
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongwei Cheng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China; Zhuhai UM Science & Technology Research Institute, University of Macau, Macau 999078, China.
| | - Gang Liu
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
7
|
Kola O, Shukla P, Haque H, Kumar A. A Single Institution Experience with a Shear-Thinning Conformable Embolic for Endovascular Embolization. Cardiovasc Intervent Radiol 2025; 48:559-566. [PMID: 40107986 PMCID: PMC11958389 DOI: 10.1007/s00270-025-04012-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE To assess the safety and efficacy of Obsidio™ conformable embolic (CE) for embolization in the peripheral vasculature. MATERIALS AND METHODS A retrospective review of the first 21 patients treated with CE was performed. Eighteen (85.7%) patients were male, and median age was 61.5 years (range, 12-89 years). Technical success was defined as stasis as assessed by a static contrast column for at least 5 heartbeats on post-embolization angiography. For procedures of peripheral vascular hemorrhage, clinical success was defined as hemorrhage resolution without reintervention within 30-day follow-up. RESULTS Indications for embolization were peripheral arterial hemorrhage (n = 13), preoperative tumor embolization (n = 4), preoperative embolization of renal cell carcinoma prior to cryoablation (n = 2), redistribution of flow prior to Yttrium-90 radioembolization to prevent nontarget radiation delivery (n = 1), and parastomal variceal embolization (n = 1). Embolization was performed via 2.4 or 2.8 French microcatheters flushed with saline prior to embolization. Most procedures (20/21) utilized < 1 cc of embolic, with the quantity used ranging between 0.1 and 1.4 cc. The amount of embolic injected was determined by the embolization endpoint, i.e., filling of the vessel intended for embolization. CE was used in combination with coils placed prior to CE in 4 procedures. Follow-up was a median of 57 days (range 0-244 days). Complete stasis was achieved in 100% (n = 21/21) of procedures. There were no post-procedure adverse events or rebleeding. CONCLUSION CE resulted in reliable vessel occlusion with no cases of rebleeding or reintervention and with no procedure-related adverse events in this series. LEVEL OF EVIDENCE Level 4, Case Series.
Collapse
Affiliation(s)
- Olivia Kola
- Rutgers - New Jersey Medical School, Newark, NJ, USA
| | - Pratik Shukla
- Rutgers - New Jersey Medical School, Newark, NJ, USA
| | - Humza Haque
- Department of Radiology, Rutgers - New Jersey Medical School, Newark, NJ, USA
| | - Abhishek Kumar
- Division of Vascular and Interventional Radiology, Department of Radiology, Rutgers - New Jersey Medical School, 185 South Orange Ave. MSB F-560, Newark, NJ, 07103, USA.
| |
Collapse
|
8
|
Shadab A, Farokhi S, Fakouri A, Mohagheghzadeh N, Noroozi A, Razavi ZS, Karimi Rouzbahani A, Zalpoor H, Mahjoor M. Hydrogel-based nanoparticles: revolutionizing brain tumor treatment and paving the way for future innovations. Eur J Med Res 2025; 30:71. [PMID: 39905470 PMCID: PMC11792566 DOI: 10.1186/s40001-025-02310-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 01/17/2025] [Indexed: 02/06/2025] Open
Abstract
Brain tumor treatment remains a significant challenge due to their high mortality and resistance to current therapies. This paper discusses the promising potential of hydrogel-based nanoparticles as innovative drug delivery systems for brain tumor therapy. Extensive characterization techniques reveal the ability of these Nano-systems to demonstrate prolonged blood circulation and targeted delivery, leading to improved survival rates. Designed with optimized physicochemical characteristics, these nanoparticles effectively cross the blood-brain barrier, circumventing a major impediment to drug delivery to the brain. By delivering drugs directly to the tumor bed, these nanoparticles enhance therapeutic outcomes and minimize adverse effects. In addition, this review investigates the techniques for characterizing, visualizing, and modifying these nanoparticles, as well as the standing challenges and promising research avenues for their clinical application. Further investigations are encouraged by this review to investigate potential advancements in hydrogel-based nanoparticle therapeutic approaches for brain tumors. This includes investigating tailored hydrogels, hybrid systems, computational modeling, and the integration of gene therapy and immunotherapy techniques. The study also addresses the need for enhanced synthesis techniques, stability, scalability, and cost-cutting measures to overcome obstacles and advance the clinical use of hydrogel-based nanoparticles in treating brain tumors.
Collapse
Affiliation(s)
- Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Deputy of Health, Iran University of Medical Sciences, Tehran, Iran
| | - Simin Farokhi
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
- USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Arshia Fakouri
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
- USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Neda Mohagheghzadeh
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Noroozi
- Dental Research Center, Faculty of Dentistry, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran
- Biochemistry Research Center, Iran University Medical Sciences, Tehran, Iran
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran
| | - Arian Karimi Rouzbahani
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
- USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
| | - Mohamad Mahjoor
- Cellular and Molecular Research Centre, Qom University of Medical Sciences, Qom, Iran.
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Wang Y, Yang J, Amier Y, Yuan D, Xun Y, Yu X. Advancements in Nanomedicine for the Diagnosis and Treatment of Kidney Stones. Int J Nanomedicine 2025; 20:1401-1423. [PMID: 39925679 PMCID: PMC11805677 DOI: 10.2147/ijn.s504318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/17/2025] [Indexed: 02/11/2025] Open
Abstract
Kidney stones constitute a common condition impacting the urinary system. In clinical diagnosis and management, traditional surgical interventions and pharmacological treatments are primarily utilized; however, these methods possess inherent limitations. Presently, the field of nanomedicine is undergoing significant advancements. The application of nanomaterials in biosensors enables the accurate assessment of urinary ion composition. Furthermore, contrast agents developed from these materials can improve the signal-to-noise ratio and enhance image clarity. By mitigating oxidative stress-induced cellular damage, nanomaterials can inhibit the formation of kidney stones and enhance the efficacy of drug delivery as effective carriers. Additionally, by modifying the physical and chemical properties of bacteria, nanomaterials can effectively eliminate bacterial presence, thereby preventing severe complications. This review explores the advancements in nanomaterials technology related to the early detection of risk factors, clinical diagnosis, and treatment of kidney stones and their associated complications.
Collapse
Affiliation(s)
- Yongqi Wang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Junyi Yang
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yirixiatijiang Amier
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Dongfeng Yuan
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yang Xun
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiao Yu
- Department of Urology, Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
10
|
Al-Asadi Z, Korona MV, Deipolyi AR. Novel Application of a Shear-Thinning Conformable Embolic Gel for Occlusion of a Bronchobiliary Fistula. Cardiovasc Intervent Radiol 2025; 48:277-279. [PMID: 39809883 DOI: 10.1007/s00270-024-03928-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025]
Affiliation(s)
- Zayd Al-Asadi
- WVU School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Michael V Korona
- Interventional Radiology, Charleston Area Medical Center, Charleston, WV, USA
| | - Amy R Deipolyi
- Interventional Radiology, WVU/Charleston Area Medical Center, 3200 MacCorkle Ave SE, Charleston, WV, 25304, USA.
| |
Collapse
|
11
|
Peng X, Tang H, Zhao Z, Zheng Y, Gui X, Jiang A, He P, Wen X, Zhang Q, Mei Z, Shi Y, Chu C, Zhang Y, Liu G. Intelligent Generic High-Throughput Oscillatory Shear Technology Fabricates Programmable Microrobots for Real-Time Visual Guidance During Embolization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2408613. [PMID: 39676403 DOI: 10.1002/smll.202408613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/07/2024] [Indexed: 12/17/2024]
Abstract
Microrobots for endovascular embolization face challenges in precise delivery within dynamic blood vessels. Here, an intelligent generic high-throughput oscillatory shear technology (iGHOST) is proposed to fabricate diversely programmable, multifunctional microrobots capable of real-time visual guidance for in vivo endovascular embolization. Leveraging machine learning (ML), key synthesis parameters affecting the success and sphericity of the microrobots are identified. Therefore, the ML-optimized iGHOST enables continuous production of uniform microrobots with programmable sizes (400-1000 µm) at an ultrahigh rate exceeding 240 mL h-1 by oscillatory segmenting fluid into droplets before ionic cross-linking, and without requiring purification. Particularly, the iGHOST-fabricated magnetically responsive lipiodol-calcium alginate (MagLiCA) microrobots are highly distinguishable under X-ray imaging, which allows for precise navigation in fluid flows of up to 4 mL min-1 and accurate embolization in liver and kidney blood vessels, thus addressing the current issues. Crucially, MagLiCA microrobots possess drug-loading capabilities, enabling simultaneous embolization and site-specific treatment. The iGHOST process is an intelligent, rapid, and green manufacturing method, which can produce size-controllable, multifunctional microrobots with the potential for precise drug delivery and treatment under real-time imaging across various medical applications.
Collapse
Affiliation(s)
- Xuqi Peng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Haitian Tang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zhenwen Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yating Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xiran Gui
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Aijun Jiang
- Department of General Surgery, Naval Medical Center of PLA, Naval Medical University, Shanghai, 200052, China
| | - Pan He
- Department of General Surgery, Institute of Hepatobiliary-Pancreatic-Intestinal Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Xiaofei Wen
- Department of Vascular & Tumor Interventional Radiology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Qian Zhang
- Institute of Artificial Intelligence, Xiamen University, Xiamen, 361102, China
| | - Ziyang Mei
- Institute of Artificial Intelligence, Xiamen University, Xiamen, 361102, China
| | - Yesi Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chengchao Chu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- Shen Zhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
12
|
Ma J, Li W, Ding Y, Chen Y, Huang X, Yu T, Song D, Niu H, Li B, Xie H, Zhang K, Yang T, Zhao X, Yang X, Ding P. Enhanced sclerotherapy for vascular malformations: A dual-mechanism approach using in-situ forming PATDs gel. Mater Today Bio 2024; 29:101376. [PMID: 39698002 PMCID: PMC11653148 DOI: 10.1016/j.mtbio.2024.101376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/13/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Vascular malformations are common vascular lesions in infants and seriously affect their health and quality of life. Vascular sclerotherapy is an effective treatment for vascular malformations. However, current sclerosants have difficulty achieving both high efficiency and low toxicity, and their dosing forms make it difficult to achieve long-term retention in the affected blood vessels. Therefore, exploring a safe and effective sclerosant and its delivery strategy is the key to clinical sclerotherapy. To address the above issues, this study developed sclerosants that could form an in-situ gel based on a dual mechanism of vascular injury and plasmin (PLA) inhibition. By linking the non-ionic surfactant sclerosant polyoxyethylene alkyl ether (PAs) and the PLA inhibitor tranexamic acid (TA) through an ester bond, a cationic surfactant sclerosant polyoxyethylene alkylether tranexamate derivatives (PATDs) were constructed. The cationic charge of PATDs enhanced its cytotoxicity to HUVEC-TIE2-L914F cells, and the ester bond of PATDs could be degraded by esterase in the blood, reducing its systemic toxicity. The degradation product TA inhibited the activation of the PLA-matrix metalloproteinase (MMPs) system induced by vascular injury, thereby promoting the deposition of collagen and the proliferation and differentiation of fibroblasts to promote vascular fibrosis. In addition, an injectable solution (PATDs/GA) was prepared by mixing PATDs with glycerol formaldehyde (GA), and PATDs/GA could form a low-molecular-weight gel automatically in an aqueous solution, which was beneficial to increase its retention in the affected blood vessels and reduce the risk of drug entering non-targeted sites. At the same time, this gel automatically dissolved, reducing the risk of immune rejection caused by long-term retention. This study provided a new and precise approach for the treatment of vascular sclerosis with high efficiency and low toxicity.
Collapse
Affiliation(s)
- Jizhuang Ma
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Wenhan Li
- Ultrasound Department, Shengjing Hospital, China Medical University, Shenyang, China
| | - Yu Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongfeng Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiaoyu Huang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Tong Yu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Di Song
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Haoran Niu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Bao Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Huichao Xie
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Keda Zhang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Tianzhi Yang
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, Husson University, Bangor, ME, USA
| | - Xiaoyun Zhao
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xinggang Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| |
Collapse
|
13
|
Chen L, Yin Q, Zhang H, Zhang J, Yang G, Weng L, Liu T, Xu C, Xue P, Zhao J, Zhang H, Yao Y, Chen X, Sun S. Protecting Against Postsurgery Oral Cancer Recurrence with an Implantable Hydrogel Vaccine for In Situ Photoimmunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309053. [PMID: 39467056 PMCID: PMC11633475 DOI: 10.1002/advs.202309053] [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: 11/23/2023] [Revised: 08/20/2024] [Indexed: 10/30/2024]
Abstract
Oral squamous cell carcinoma (OSCC) often recurs aggressively and metastasizes despite surgery and adjuvant therapy, driven by postoperative residual cancer cells near the primary tumor site. An implantable in situ vaccine hydrogel was designed to target residual OSCC cells post-tumor removal. This hydrogel serves as a reservoir for the sustained localized release of δ-aminolevulinic acid (δ-ALA), enhancing protoporphyrin IX-mediated photodynamic therapy (PDT), and a polydopamine-hyaluronic acid composite for photothermal therapy (PTT). Additionally, immune adjuvants, including anti-CD47 antibodies (aCD47) and CaCO3 nanoparticles, are directly released into the resected tumor bed. This approach induces apoptosis of residual OSCC cells through sequential near-infrared irradiation, promoting calcium interference therapy (CIT). The hydrogel further stimulates immunogenic cell death (ICD), facilitating the polarization of tumor-associated macrophages from the M2 to the M1 phenotype. This facilitates phagocytosis, dendritic cell activation, robust antigen presentation, and cytotoxic T lymphocyte-mediated cytotoxicity. In murine OSCC models, the in situ vaccine effectively prevents local recurrence, inhibits orthotopic OSCC growth and pulmonary metastases, and provides long-term protective immunity against tumor rechalle nge. These findings support postoperative in situ vaccination with a biocompatible hydrogel implant as a promising strategy to minimize residual tumor burden and reduce recurrence risk after OSCC resection.
Collapse
Affiliation(s)
- Lan Chen
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Qiqi Yin
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Handan Zhang
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Jie Zhang
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Guizhu Yang
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Lin Weng
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Tao Liu
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Chenghui Xu
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Pengxin Xue
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Jinchao Zhao
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Han Zhang
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Yanli Yao
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| | - Xin Chen
- School of Chemical Engineering and TechnologyShaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical EngineeringXi'an Jiaotong UniversityXi'an710049China
| | - Shuyang Sun
- Department of Oral and Maxillofacial‐Head Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai Research Institute of StomatologyResearch Unit of Oral and Maxillofacial Regenerative MedicineChinese Academy of Medical SciencesShanghai200011China
| |
Collapse
|
14
|
Yu Q, Ahmed O, Chen J, Islam Y, Funaki B, Patel M. A novel conformable embolic for selective transarterial embolization of acute hemorrhages: a technical note. CVIR Endovasc 2024; 7:78. [PMID: 39503974 PMCID: PMC11541968 DOI: 10.1186/s42155-024-00492-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 10/22/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Obsidio conformable embolic (OCE, Boston Scientific, MA) is a novel, radiopaque and conformable embolic. The purpose of this report is to describe its use for treatment of acute intra-abdominal hemorrhages. METHODS AND RESULTS Three patients presented with acute hemorrhage and were treated with OCE, including post-paracentesis hemorrhage, penetrating trauma to the liver, and blunt trauma in the spleen. All cases were performed under moderate sedation, with hemostasis achieved by end of procedure using less than 1 vial of OCE (0.2-0.4 ml). No severe adverse events occurred. None required repeated treatment. SHORT CONCLUSION OCE is a safe and effective embolic agent for treatment of intra-abdominal or visceral hemorrhage. Future studies with larger sample sizes and longer follow-up are warranted.
Collapse
Affiliation(s)
- Qian Yu
- Department of Radiology, University of Chicago Medical Center, University of Chicago, Chicago, IL, 60637, USA.
| | - Osman Ahmed
- Department of Radiology, University of Chicago Medical Center, University of Chicago, Chicago, IL, 60637, USA
| | - Jiaqi Chen
- Chicago Medical School, 3333 N Green Bay Rd, North Chicago, IL, 60064, USA
| | - Yousuf Islam
- Department of Radiology, University of Chicago Medical Center, University of Chicago, Chicago, IL, 60637, USA
| | - Brian Funaki
- Department of Radiology, University of Chicago Medical Center, University of Chicago, Chicago, IL, 60637, USA
| | - Mikin Patel
- Department of Radiology, University of Chicago Medical Center, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
15
|
Albadawi H, Zhang Z, Keum H, Cevik E, Nagalo BM, Gunduz S, Kita H, Oklu R. Percutaneous Delivery of Oncogel for Targeted Liver Tumor Ablation and Controlled Release of Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406080. [PMID: 39148179 PMCID: PMC11543513 DOI: 10.1002/adma.202406080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/30/2024] [Indexed: 08/17/2024]
Abstract
Advanced-stage liver cancers are associated with poor prognosis and have limited treatment options, often leading the patient to hospice care. Percutaneous intratumoral injection of anticancer agents has emerged as a potential alternative to systemic therapy to overcome tumor barriers, increase bioavailability, potentiate immunotherapy, and avoid systemic toxicity, which advanced-stage cancer patients cannot tolerate. Here, an injectable OncoGel (OG) comprising of a nanocomposite hydrogel loaded with an ionic liquid (IL) is developed for achieving a predictable and uniform tumor ablation and long-term slow release of anticancer agents into the ablation zone. Rigorous mechanical, physiochemical, drug release, cytotoxicity experiments, and ex vivo human tissue testing identify an injectable version of the OG with bactericidal properties against highly resistant bacteria. Intratumoral injection of OG loaded with Nivolumab (Nivo) and doxorubicin (Dox) into highly malignant tumor models in mice, rats, and rabbits demonstrates enhanced survival and tumor regression associated with robust tissue ablation and drug distribution throughout the tumor. Mass cytometry and proteomic studies in a mouse model of colorectal cancer that often metastasizes to the liver indicate an enhanced anticancer immune response following the intratumoral injection of OG. OG may augment immunotherapy and potentially improve outcomes in liver cancer patients.
Collapse
Affiliation(s)
- Hassan Albadawi
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Zefu Zhang
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Hyeongseop Keum
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Enes Cevik
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Bolni M Nagalo
- University of Arkansas for Medical Sciences, College of Medicine, Department of Pathology, 301 West Markham Street, Little Rock, AR, 72205, USA
| | - Seyda Gunduz
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
- Department of Medical Oncology, Istinye University; Bahcesehir Liv Hospital, Istanbul 34517, Turkey
| | - Hirohito Kita
- Department of Immunology, Division of Allergy, Asthma, and Clinical Immunology and the Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona 85259, USA
| | - Rahmi Oklu
- Division of Vascular & Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| |
Collapse
|
16
|
Chen Y, Li C, Yang J, Wang M, Wang Y, Cheng S, Huang W, Yuan G, Xie M. Intravascular elimination of circulating tumor cells and cascaded embolization with multifunctional 3D tubular scaffolds. J Mater Chem B 2024; 12:9018-9029. [PMID: 39158001 DOI: 10.1039/d4tb01151a] [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: 08/20/2024]
Abstract
The primary tumor ("root") and circulating tumor cells (CTCs; "seeds") are vital factors in tumor progression. However, current treatment strategies mainly focus on inhibiting the tumor while ignoring CTCs, resulting in tumor metastasis. Here, we design a multifunctional 3D scaffold with interconnected macropores, excellent photothermal ability and perfect bioaffinity as a blood vessel implantable device. When implanted upstream of the primary tumor, the scaffold intercepts CTCs fleeing back to the primary tumor and then forms "micro-thrombi" to block the supply of nutrients and oxygen to the tumor for embolization therapy. The scaffold implanted downstream of the tumor efficiently captures and photothermally kills the CTCs that escape from the tumor, thereby preventing metastasis. Experiments using rabbits demonstrated excellent biosafety of this scaffold with 86% of the CTC scavenging rate, 99% of the tumor inhibition rate and 100% of CTC killing efficiency. The multifunctional 3D scaffold synergistically inhibits the "root" and eliminates the "seeds" of the tumor, demonstrating its potential for localized cancer therapy with few side effects and high antitumor efficacy.
Collapse
Affiliation(s)
- Yijing Chen
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Cuiwen Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Jinghui Yang
- School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Ming Wang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yike Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Shibo Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Weihua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Guohua Yuan
- School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Min Xie
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
17
|
Enayati M, Liu W, Madry H, Neisiany RE, Cucchiarini M. Functionalized hydrogels as smart gene delivery systems to treat musculoskeletal disorders. Adv Colloid Interface Sci 2024; 331:103232. [PMID: 38889626 DOI: 10.1016/j.cis.2024.103232] [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: 01/15/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Despite critical advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy based on the delivery of therapeutic genetic sequences has strong value to offer effective, durable options to decisively manage such disorders. Furthermore, scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy, allowing for the spatiotemporal delivery of candidate genes to sites of injury. Among the many scaffolds for musculoskeletal research, hydrogels raised increasing attention in addition to other potent systems (solid, hybrid scaffolds) due to their versatility and competence as drug and cell carriers in tissue engineering and wound dressing. Attractive functionalities of hydrogels for musculoskeletal therapy include their injectability, stimuli-responsiveness, self-healing, and nanocomposition that may further allow to upgrade of them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. Such functionalized hydrogels may also be tuned to successfully transfer therapeutic genes in a minimally invasive manner in order to protect their cargos and allow for their long-term effects. In light of such features, this review focuses on functionalized hydrogels and demonstrates their competence for the treatment of musculoskeletal disorders using gene therapy procedures, from gene therapy principles to hydrogel functionalization methods and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are being discussed in the perspective of translation in patients. STATEMENT OF SIGNIFICANCE: Despite advances in regenerative medicine, the generation of definitive, reliable treatments for musculoskeletal diseases remains challenging. Gene therapy has strong value in offering effective, durable options to decisively manage such disorders. Scaffold-mediated gene therapy provides powerful alternatives to overcome hurdles associated with classical gene therapy. Among many scaffolds for musculoskeletal research, hydrogels raised increasing attention. Functionalities including injectability, stimuli-responsiveness, and self-healing, tune them as "intelligently" efficient and mechanically strong platforms, rather than as just inert vehicles. This review introduces functionalized hydrogels for musculoskeletal disorder treatment using gene therapy procedures, from gene therapy principles to functionalized hydrogels and applications of hydrogel-mediated gene therapy for musculoskeletal disorders, while remaining challenges are discussed from the perspective of translation in patients.
Collapse
Affiliation(s)
- Mohammadsaeid Enayati
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany
| | - Rasoul Esmaeely Neisiany
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, 44-100 Gliwice, Poland; Department of Polymer Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, 66421 Homburg, Saar, Germany.
| |
Collapse
|
18
|
Keum H, Kim J, Zhang Z, Graf E, Albadawi H, Oklu R. Biocompatible Liquid Embolic for the Treatment of Microvascular Hemorrhage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403615. [PMID: 39049735 PMCID: PMC11425845 DOI: 10.1002/advs.202403615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/25/2024] [Indexed: 07/27/2024]
Abstract
Persistent or recurrent bleeding from microvessels inaccessible for direct endovascular intervention is a major problem in medicine today. Here, an innovative catheter-directed liquid embolic (P-LE) is bioengineered for rapid microvessel embolization to treat small vessel hemorrhage. Tested in rodent, porcine, and canine animal models under normal and coagulopathic conditions, P-LE outperformed clinically used embolic materials in both survival and non-survival experiments, effectively occluding vessels as small as 40 microns with no signs of recanalization. P-LE occlusion is independent of the coagulation cascade, and its resistance to displacement is ≈ 8 times greater than systolic blood pressure. P-LE is also found to be biocompatible and x-ray visible and does not require polymerization or a chemical reaction to embolize. To simulate the clinical scenario, acute microvascular hemorrhage is created in the pig kidney, liver, or stomach; these are successfully treated with P-LE achieving immediate hemostasis. Furthermore, P-LE is found to be bactericidal to highly resistant patient-derived bacteria, suggesting that P-LE may also protect against infectious complications that may occur following embolization procedures. P-LE is safe, easy to use, and effective in treating -microvessel hemorrhage.
Collapse
Affiliation(s)
- Hyeongseop Keum
- The Laboratory for Patient‐Inspired Engineering, Mayo Clinic13400 East Shea BlvdScottsdaleAZ85259USA
| | - Jinjoo Kim
- The Laboratory for Patient‐Inspired Engineering, Mayo Clinic13400 East Shea BlvdScottsdaleAZ85259USA
| | - Zefu Zhang
- The Laboratory for Patient‐Inspired Engineering, Mayo Clinic13400 East Shea BlvdScottsdaleAZ85259USA
| | - Erin Graf
- Department of Laboratory Medicine and Pathology, Mayo Clinic5777 E Mayo BlvdPhoenixAZ85054USA
| | - Hassan Albadawi
- The Laboratory for Patient‐Inspired Engineering, Mayo Clinic13400 East Shea BlvdScottsdaleAZ85259USA
- Division of Vascular & Interventional Radiology, Mayo Clinic5777 E Mayo BlvdPhoenixAZ85054USA
| | - Rahmi Oklu
- The Laboratory for Patient‐Inspired Engineering, Mayo Clinic13400 East Shea BlvdScottsdaleAZ85259USA
- Division of Vascular & Interventional Radiology, Mayo Clinic5777 E Mayo BlvdPhoenixAZ85054USA
| |
Collapse
|
19
|
Kim S, Nowicki KW, Kohyama K, Mittal A, Ye S, Wang K, Fujii T, Rajesh S, Cao C, Mantena R, Barbuto M, Jung Y, Gross BA, Friedlander RM, Wagner WR. Development of an Injectable, ECM-Derivative Embolic for the Treatment of Cerebral Saccular Aneurysms. Biomacromolecules 2024; 25:4879-4890. [PMID: 39001820 PMCID: PMC11323012 DOI: 10.1021/acs.biomac.4c00321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
Cerebral aneurysms are a source of neurological morbidity and mortality, most often as a result of rupture. The most common approach for treating aneurysms involves endovascular embolization using nonbiodegradable medical devices, such as platinum coils. However, the need for retreatment due to the recanalization of coil-treated aneurysms highlights the importance of exploring alternative solutions. In this study, we propose an injectable extracellular matrix-derived embolic formed in situ by Michael addition of gelatin-thiol (Gel-SH) and hyaluronic acid vinyl sulfone (HA-VS) that may be delivered with a therapeutic agent (here, RADA-SP) to fill and remodel aneurysmal tissue without leaving behind permanent foreign bodies. The injectable embolic material demonstrated rapid gelation under physiological conditions, forming a highly porous structure and allowing for cellular infiltration. The injectable embolic exhibited thrombogenic behavior in vitro that was comparable to that of alginate injectables. Furthermore, in vivo studies in a murine carotid aneurysm model demonstrated the successful embolization of a saccular aneurysm and extensive cellular infiltration both with and without RADA-SP at 3 weeks, with some evidence of increased vascular or fibrosis markers with RADA-SP incorporation. The results indicate that the developed embolic has inherent potential for acutely filling cerebrovascular aneurysms and encouraging the cellular infiltration that would be necessary for stable, chronic remodeling.
Collapse
Affiliation(s)
- Seungil Kim
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department
of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- McGowan
Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Kamil W. Nowicki
- Department
of Neurosurgery, School of Medicine, University
of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department
of Neurosurgery, School of Medicine, Yale, New
Haven, Connecticut 06520, United States
| | - Keishi Kohyama
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- McGowan
Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Aditya Mittal
- Department
of Neurosurgery, School of Medicine, University
of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Sangho Ye
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department
of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- McGowan
Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Kai Wang
- Discovery
Center for Musculoskeletal Recovery, Schoen
Adams Research Institute at Spaulding, Charlestown, Massachusetts 02115, United States
- Department
of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Taro Fujii
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- McGowan
Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Shivbaskar Rajesh
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- McGowan
Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Catherine Cao
- Division
of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
| | - Rohit Mantena
- Department
of Neurosurgery, School of Medicine, University
of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Marianna Barbuto
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department
of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- McGowan
Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Ri.MED
Foundation, Cardiac Tissue Engineering Laboratory, Ri.MED Foundation, Palermo 90133, Italy
- Department
of Biological, Chemical and Pharmaceutical
Sciences and Technologies (STEBICEF), University of Palermo, Palermo 90133, Italy
| | - Youngmee Jung
- Center
for Biomaterials, Biomedical Research Institute, Korea Institute of
Science and Technology (KIST), Seoul 130-650, Republic
of Korea
- School of
Electrical and Electronic Engineering, YU-KIST
Institute, Yonsei University, Seoul 130-650 Republic of Korea
| | - Bradley A. Gross
- Department
of Neurosurgery, School of Medicine, University
of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Robert M. Friedlander
- Department
of Neurosurgery, School of Medicine, University
of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - William R. Wagner
- Department
of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- Department
of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
- McGowan
Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
20
|
Pal K, Patel M, Chen SR, Odisio BC, Metwalli Z, Ahrar J, Irwin D, Sheth RA, Kuban JD. A Single-Center Experience with a Shear-Thinning Conformable Embolic. J Vasc Interv Radiol 2024; 35:1215-1220. [PMID: 38719091 DOI: 10.1016/j.jvir.2024.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 06/07/2024] Open
Abstract
The purpose of this study was to evaluate the technical success, effectiveness, and safety of transarterial embolization for acute bleeding management with a shear-thinning conformable embolic. This single-center retrospective study evaluated outcomes after embolization using Obsidio conformable embolic (OCE). Technical success was defined as performing transarterial embolization within the target vessel to complete stasis of antegrade flow. Treatment effectiveness was defined as cessation of bleeding for patients. Eleven patients underwent 11 embolization procedures. A total of 16 arteries were embolized. Indications for embolization were spontaneous tumor bleeding (6/11), hematuria (2/11), active duodenal bleeding (1/11), portal hypertensive bleeding (1/11), and rectus sheath hematoma (1/11). The technical success rate was 100%. The median vessel diameter was 2 mm (range, 1-3 mm). There were no adverse events or off-target embolization. OCE demonstrated technical success and treatment effectiveness with a short-term safety profile for transarterial embolization interventions.
Collapse
Affiliation(s)
- Koustav Pal
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Milan Patel
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephen R Chen
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bruno C Odisio
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zeyad Metwalli
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Judy Ahrar
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Irwin
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rahul A Sheth
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joshua D Kuban
- Department of Interventional Radiology, the University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
21
|
Lin X, Liu R, Beitzel J, Zhou Y, Lagadon C, Zhang M. Injectable Biodegradable Chitosan-PEG/PEG-Dialdehyde Hydrogel for Stem Cell Delivery and Cartilage Regeneration. Gels 2024; 10:508. [PMID: 39195037 DOI: 10.3390/gels10080508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 08/29/2024] Open
Abstract
Stem cell-based therapy holds promise for cartilage regeneration in treating knee osteoarthritis (KOA). Injectable hydrogels have been developed to mimic the extracellular matrix (ECM) and facilitate stem cell growth, proliferation, and differentiation. However, these hydrogels face limitations such as poor mechanical strength, inadequate biocompatibility, and suboptimal biodegradability, collectively hindering their effectiveness in cartilage regeneration. This study introduces an injectable, biodegradable, and self-healing hydrogel composed of chitosan-PEG and PEG-dialdehyde for stem cell delivery. This hydrogel can form in situ by blending two polymer solutions through injection at physiological temperature, encapsulating human adipose-derived stem cells (hADSCs) during the gelation process. Featuring a 3D porous structure with large pore size, optimal mechanical properties, biodegradability, easy injectability, and rapid self-healing capability, the hydrogel supports the growth, proliferation, and differentiation of hADSCs. Notably, encapsulated hADSCs form 3D spheroids during proliferation, with their sizes increasing over time alongside hydrogel degradation while maintaining high viability for at least 10 days. Additionally, hADSCs encapsulated in this hydrogel exhibit upregulated expression of chondrogenic differentiation genes and proteins compared to those cultured on 2D surfaces. These characteristics make the chitosan-PEG/PEG-dialdehyde hydrogel-stem cell construct suitable for direct implantation through minimally invasive injection, enhancing stem cell-based therapy for KOA and other cell-based treatments.
Collapse
Affiliation(s)
- Xiaojie Lin
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Ruofan Liu
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Jacob Beitzel
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Chloe Lagadon
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
22
|
Yu C, Qiu Y, Yao F, Wang C, Li J. Chemically Programmed Hydrogels for Spatiotemporal Modulation of the Cardiac Pathological Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404264. [PMID: 38830198 DOI: 10.1002/adma.202404264] [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: 03/24/2024] [Revised: 05/20/2024] [Indexed: 06/05/2024]
Abstract
After myocardial infarction (MI), sustained ischemic events induce pathological microenvironments characterized by ischemia-hypoxia, oxidative stress, inflammatory responses, matrix remodeling, and fibrous scarring. Conventional clinical therapies lack spatially targeted and temporally responsive modulation of the infarct microenvironment, leading to limited myocardial repair. Engineered hydrogels have a chemically programmed toolbox for minimally invasive localization of the pathological microenvironment and personalized responsive modulation over different pathological periods. Chemically programmed strategies for crosslinking interactions, interfacial binding, and topological microstructures in hydrogels enable minimally invasive implantation and in situ integration tailored to the myocardium. This enhances substance exchange and signal interactions within the infarcted microenvironment. Programmed responsive polymer networks, intelligent micro/nanoplatforms, and biological therapeutic cues contribute to the formation of microenvironment-modulated hydrogels with precise targeting, spatiotemporal control, and on-demand feedback. Therefore, this review summarizes the features of the MI microenvironment and chemically programmed schemes for hydrogels to conform, integrate, and modulate the cardiac pathological microenvironment. Chemically programmed strategies for oxygen-generating, antioxidant, anti-inflammatory, provascular, and electrointegrated hydrogels to stimulate iterative and translational cardiac tissue engineering are discussed.
Collapse
Affiliation(s)
- Chaojie Yu
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Yuwei Qiu
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Fanglian Yao
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Changyong Wang
- Tissue Engineering Research Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Junjie Li
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| |
Collapse
|
23
|
Gao X, Caruso BR, Li W. Advanced Hydrogels in Breast Cancer Therapy. Gels 2024; 10:479. [PMID: 39057502 PMCID: PMC11276203 DOI: 10.3390/gels10070479] [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: 07/01/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is the most common malignancy among women and is the second leading cause of cancer-related death for women. Depending on the tumor grade and stage, breast cancer is primarily treated with surgery and antineoplastic therapy. Direct or indirect side effects, emotional trauma, and unpredictable outcomes accompany these traditional therapies, calling for therapies that could improve the overall treatment and recovery experiences of patients. Hydrogels, biomimetic materials with 3D network structures, have shown great promise for augmenting breast cancer therapy. Hydrogel implants can be made with adipogenic and angiogenic properties for tissue integration. 3D organoids of malignant breast tumors grown in hydrogels retain the physical and genetic characteristics of the native tumors, allowing for post-surgery recapitulation of the diseased tissues for precision medicine assessment of the responsiveness of patient-specific cancers to antineoplastic treatment. Hydrogels can also be used as carrier matrices for delivering chemotherapeutics and immunotherapeutics or as post-surgery prosthetic scaffolds. The hydrogel delivery systems could achieve localized and controlled medication release targeting the tumor site, enhancing efficacy and minimizing the adverse effects of therapeutic agents delivered by traditional procedures. This review aims to summarize the most recent advancements in hydrogel utilization for breast cancer post-surgery tissue reconstruction, tumor modeling, and therapy and discuss their limitations in clinical translation.
Collapse
Affiliation(s)
- Xiangyu Gao
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Benjamin R. Caruso
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
24
|
Zhu R, Wang R, Li J, Chen M, Qiu L, Bai S. An artificial liquid-liquid phase separation-driven silk fibroin-based adhesive for rapid hemostasis and wound sealing. Acta Biomater 2024; 182:14-27. [PMID: 38750918 DOI: 10.1016/j.actbio.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/24/2024]
Abstract
The powerful adhesion systems of marine organisms have inspired the development of artificial protein-based bioadhesives. However, achieving robust wet adhesion using artificial bioadhesives remains technically challenging because the key element of liquid-liquid phase separation (LLPS)-driven complex coacervation in natural adhesion systems is often ignored. In this study, mimicking the complex coacervation phenomenon of marine organisms, an artificial protein-based adhesive hydrogel (SFG hydrogel) was developed by adopting the LLPS-mediated coacervation of the natural protein silk fibroin (SF) and the anionic surfactant sodium dodecylbenzene sulfonate (SDBS). The assembled SF/SDBS complex coacervate enabled precise spatial positioning and easy self-adjustable deposition on irregular substrate surfaces, allowing for tight contact. Spontaneous liquid-to-solid maturation promoted the phase transition of the SF/SDBS complex coacervate to form the SFG hydrogel in situ, enhancing its bulk cohesiveness and interfacial adhesion. The formed SFG hydrogel exhibited intrinsic advantages as a new type of artificial protein-based adhesive, including good biocompatibility, robust wet adhesion, rapid blood-clotting capacity, and easy operation. In vitro and in vivo experiments demonstrated that the SFG hydrogel not only achieved instant and effective hemostatic sealing of tissue injuries but also promoted wound healing and tissue regeneration, thus advancing its clinical applications. STATEMENT OF SIGNIFICANCE: Marine mussels utilize the liquid-liquid phase separation (LLPS) strategy to induce the supramolecular assembly of mussel foot proteins, which plays a critical role in strong underwater adhesion of mussel foot proteins. Herein, an artificial protein-based adhesive hydrogel (named SFG hydrogel) was reported by adopting the LLPS-mediated coacervation of natural protein silk fibroin (SF) and anionic surfactant sodium dodecylbenzene sulfonate (SDBS). The assembled SFG hydrogel enabled the precise spatial positioning and easy self-adjustable deposition on substrate surfaces with irregularities, allowing tight interfacial adhesion and cohesiveness. The SFG hydrogel not only achieved instant and effective hemostatic sealing of tissue injuries but also promoted wound healing and tissue regeneration, exhibiting intrinsic advantages as a new type of artificial protein-based bioadhesives.
Collapse
Affiliation(s)
- Rui Zhu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Ruiheng Wang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Jie Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Minghui Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Lingyu Qiu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Shumeng Bai
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, People's Republic of China.
| |
Collapse
|
25
|
Tao S, Zhu S, Wang W, Cao X, Hu Y, Chen Q, Zha L, Zha Z. Shape Self-Adaptive Liquid Embolic Agent for Ultrafast and Durable Vascular Embolization. ACS APPLIED MATERIALS & INTERFACES 2024; 16:31936-31949. [PMID: 38869429 DOI: 10.1021/acsami.4c02892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Minimally invasive embolization greatly decreases the mortality resulting from vascular injuries while still suffering from a high risk of recanalization and systematic thrombosis due to the intrinsic hydrophobicity and poor adhesion of the clinically used liquid embolic agent of Lipiodol. In this study, a shape self-adaptive liquid embolic agent was developed by mixing biocompatible poly(acrylic acid) (PAA), two-dimensional magnesium-aluminum layered double hydroxide (LDH), and poly(ethylene glycol)200 (PEG200). Upon contact with blood, the injectable PAA-LDH@PEG200 would quickly absorb water to form an adhesive and mechanically strong PAA-LDH thin hydrogel within 5 s, which could firmly adhere to the blood vessel wall for ultrafast and durable embolization. In addition, benefiting from the "positively charged nucleic center effect" of LDH nanosheets, the liquid PAA-LDH@PEG200 could avoid vascular distension by PAA overexpansion and possess high shock-resistant mechanical strength from the blood flow. Furthermore, both in vitro and in vivo embolization experiments demonstrated the complete embolic capacity of liquid PAA-LDH@PEG200 without the occurrence of recanalization for 28 days and also the great potential to act as a platform to couple with chemotherapeutic drugs for the minimized transcatheter arterial chemoembolization (TACE) treatment of VX2 tumors without recurrence for 18 days. Thus, liquid PAA-LDH@PEG200 developed here possesses great potential to act as a shape self-adaptive liquid embolic agent for ultrafast and durable vascular embolization.
Collapse
Affiliation(s)
- Shi Tao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Shuangli Zhu
- Institute of Medical Health, Harbin Institute of Technology Zhengzhou Research Institute, Zhengzhou 450000, P. R. China
| | - Weitao Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Xiangjing Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Yaoyu Hu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Qian Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Lisha Zha
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, P. R. China
- School of Biomedical Sciences, Hunan University, Changsha 410082, P. R. China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| |
Collapse
|
26
|
Wu WS, Yan X, Chen S, Du Y, Hu J, Song Y, Zha Z, Xu YJ, Cao B, Xuan SH, Liu X, Chen B, Dong L, Lu Y, Yu SH. Minimally Invasive Delivery of Percutaneous Ablation Agent via Magnetic Colloidal Hydrogel Injection for Treatment of Hepatocellular Carcinoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309770. [PMID: 38447017 DOI: 10.1002/adma.202309770] [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: 09/20/2023] [Revised: 02/28/2024] [Indexed: 03/08/2024]
Abstract
Percutaneous thermotherapy, a minimally invasive operational procedure, is employed in the ablation of deep tumor lesions by means of target-delivering heat. Conventional thermal ablation methods, such as radiofrequency or microwave ablation, to a certain extent, are subjected to extended ablation time as well as biosafety risks of unwanted overheating. Given its effectiveness and safety, percutaneous thermotherapy gains a fresh perspective, thanks to magnetic hyperthermia. In this respect, an injectable- and magnetic-hydrogel-construct-based thermal ablation agent is likely to be a candidate for the aforementioned clinical translation. Adopting a simple and environment-friendly strategy, a magnetic colloidal hydrogel injection is introduced by a binary system comprising super-paramagnetic Fe3O4 nanoparticles and gelatin nanoparticles. The colloidal hydrogel constructs, unlike conventional bulk hydrogel, can be easily extruded through a percutaneous needle and then self-heal in a reversible manner owing to the unique electrostatic cross-linking. The introduction of magnetic building blocks is exhibited with a rapid magnetothermal response to an alternating magnetic field. Such hydrogel injection is capable of generating heat without limitation of deep penetration. The materials achieve outstanding therapeutic results in mouse and rabbit models. These findings constitute a new class of locoregional interventional thermal therapies with minimal collateral damages.
Collapse
Affiliation(s)
- Wen-Shu Wu
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xu Yan
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Sheng Chen
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Yaxin Du
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Jinlong Hu
- Department of General Surgery, Department of Ultrasonics, Department of Interventional Radiology, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui, 230041, P. R. China
| | - Yonghong Song
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Zhengbao Zha
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Yun-Jun Xu
- New Cornerstone Science Laboratory, Division of Nanomaterials and Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei, 230026, China
| | - Baoqiang Cao
- Department of General Surgery, Department of Ultrasonics, Department of Interventional Radiology, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui, 230041, P. R. China
| | - Shou-Hu Xuan
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, CAS Center for Excellence in Complex System Mechanics, University of Science and Technology of China, Hefei, 230027, China
| | - Xingyu Liu
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Bing Chen
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Liang Dong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yang Lu
- Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Shu-Hong Yu
- New Cornerstone Science Laboratory, Division of Nanomaterials and Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
27
|
George VPJ, Zhao K, Chen P, Hu J. Chitosan-nanoclay embolic material for catheter-directed arterial embolization. J Biomed Mater Res A 2024; 112:914-930. [PMID: 38229508 PMCID: PMC10984788 DOI: 10.1002/jbm.a.37670] [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: 08/25/2023] [Revised: 11/21/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Minimally invasive transcatheter embolization is a common nonsurgical procedure in interventional radiology. It is used for the deliberate occlusion of blood vessels for the treatment of disease or injured vasculature, including vascular malformation and malignant/benign tumors. Here, we introduce a gel embolic agent comprising chitosan nanofibers and nanoclay with excellent catheter injectability and tunable mechanical properties for embolization. The properties of the gel were optimized by varying the ratio between each individual component and also adjusting the total solid content. The rheological studies confirm the shear thinning property and gel nature of the developed gel as well as their recoverability. Injection force was measured to record the force required to pass the embolic gel through a clinically relevant catheter, evaluating for practicality of hand-injection. Theoretical predicted injection force was calculated to reduce the development time and to enhance the physician's experience. The stability of occlusion was also tested in vitro by monitoring the pressure required to displace the gel. The engineered gels exhibited sterility, hemocompatibility and cell biocompatibility, highlighting their potential for transcatheter embolization.
Collapse
Affiliation(s)
- Varghese P J George
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC, USA
| | - Keren Zhao
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC, USA
| | - Peng Chen
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC, USA
| | - Jingjie Hu
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
28
|
Perri P, Sena G, Piro P, De Bartolo T, Galassi S, Costa D, Serra R. Onyx TMGel or Coil versus Hydrogel as Embolic Agents in Endovascular Applications: Review of the Literature and Case Series. Gels 2024; 10:312. [PMID: 38786229 PMCID: PMC11120993 DOI: 10.3390/gels10050312] [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: 03/27/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
This review focuses on the use of conventional gel or coil and "new" generation hydrogel used as an embolic agent in endovascular applications. In general, embolic agents have deep or multidistrict vascular penetration properties as they ensure complete occlusion of vessels by exploiting the patient's coagulation system, which recognises them as substances foreign to the body, thus triggering the coagulation cascade. This is why they are widely used in the treatment of endovascular corrections (EV repair), arteriovenous malformations (AVM), endoleaks (E), visceral aneurysms or pseudo-aneurysms, and embolisation of pre-surgical or post-surgical (iatrogenic) lesions. Conventional gels such as Onyx or coils are now commercially available, both of which are frequently used in endovascular interventional procedures, as they are minimally invasive and have numerous advantages over conventional open repair (OR) surgery. Recently, these agents have been modified and optimised to develop new embolic substances in the form of hydrogels based on alginate, chitosan, fibroin and other polymers to ensure embolisation through phase transition phenomena. The main aim of this work was to expand on the data already known in the literature concerning the application of these devices in the endovascular field, focusing on the advantages, disadvantages and safety profiles of conventional and innovative embolic agents and also through some clinical cases reported. The clinical case series concerns the correction and exclusion of endoleak type I or type II appeared after an endovascular procedure of exclusion of aneurysmal abdominal aortic (EVAR) with a coil (coil penumbra released by a LANTERN microcatheter), the exclusion of renal arterial malformation (MAV) with a coil (penumbra coil released by a LANTERN microcatheter) and the correction of endoleak through the application of Onyx 18 in the arteries where sealing by the endoprosthesis was not guaranteed.
Collapse
Affiliation(s)
- Paolo Perri
- Department of Vascular and Endovascular Surgery, Annunziata Hospital, 1 Via Migliori, 87100 Cosenza, Italy; (P.P.); (P.P.)
| | - Giuseppe Sena
- Department of Vascular Surgery, “Pugliese-Ciaccio” Hospital, 88100 Catanzaro, Italy;
| | - Paolo Piro
- Department of Vascular and Endovascular Surgery, Annunziata Hospital, 1 Via Migliori, 87100 Cosenza, Italy; (P.P.); (P.P.)
| | - Tommaso De Bartolo
- Departement of Interventional Radiology, Annunziata Hospital, 1 Via Migliori, 87100 Cosenza, Italy; (T.D.B.); (S.G.)
| | - Stefania Galassi
- Departement of Interventional Radiology, Annunziata Hospital, 1 Via Migliori, 87100 Cosenza, Italy; (T.D.B.); (S.G.)
| | - Davide Costa
- Interuniversity Center of Phlebolymphology (CIFL), Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
29
|
Fu W, Guo M, Zhou X, Wang Z, Sun J, An Y, Guan T, Hu M, Li J, Chen Z, Ye J, Gao X, Gao GF, Dai L, Wang Y, Chen C. Injectable Hydrogel Mucosal Vaccine Elicits Protective Immunity against Respiratory Viruses. ACS NANO 2024; 18:11200-11216. [PMID: 38620102 DOI: 10.1021/acsnano.4c00155] [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: 04/17/2024]
Abstract
Intranasal vaccines, eliciting mucosal immune responses, can prevent early invasion, replication, and transmission of pathogens in the respiratory tract. However, the effective delivery of antigens through the nasal barrier and boosting of a robust systematic and mucosal immune remain challenges in intranasal vaccine development. Here, we describe an intranasally administered self-healing hydrogel vaccine with a reversible strain-dependent sol-gel transition by precisely modulating the self-assembly processes between the natural drug rhein and aluminum ions. The highly bioadhesive hydrogel vaccine enhances antigen stability and prolongs residence time in the nasal cavity and lungs by confining the antigen to the surface of the nasal mucosa, acting as a "mucosal mask". The hydrogel also stimulates superior immunoenhancing properties, including antigen internalization, cross-presentation, and dendritic cell maturation. Furthermore, the formulation recruits immunocytes to the nasal mucosa and nasal-associated lymphoid tissue (NALT) while enhancing antigen-specific humoral, cellular, and mucosal immune responses. Our findings present a promising strategy for preparing intranasal vaccines for infectious diseases or cancer.
Collapse
Affiliation(s)
- Wenjiao Fu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Xuemei Zhou
- School of Life Sciences, Hebei University, Baoding 071002, People's Republic of China
| | - Zhenzhen Wang
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Jiufeng Sun
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, People's Republic of China
| | - Yaling An
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Tong Guan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Mingdi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jiayang Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Ziwei Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Jinmin Ye
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Xingfa Gao
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Lianpan Dai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Yaling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100021, People's Republic of China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100021, People's Republic of China
| |
Collapse
|
30
|
Ai S, Gao Q, Cheng G, Zhong P, Cheng P, Ren Y, Wang H, Zhu X, Guan S, Qu X. Construction of an Injectable Composite Double-Network Hydrogel as a Liquid Embolic Agent. Biomacromolecules 2024; 25:2052-2064. [PMID: 38426456 DOI: 10.1021/acs.biomac.3c01437] [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: 03/02/2024]
Abstract
Conventional embolists disreputably tend to recanalization arising from the low filling ratio due to their rigidity or instability. As a result, intelligent hydrogels with a tunable modulus may meaningfully improve the therapeutic efficacy. Herein, an injectable composite double-network (CDN) hydrogel with high shear responsibility was prepared as a liquid embolic agent by cross-linking poly(vinyl alcohol) (PVA) and carboxymethyl chitosan (CMC) via dynamic covalent bonding of borate ester and benzoic-imine. A two-dimensional nanosheet, i.e., layered double hydroxide (LDH), was incorporated into the network through physical interactions which led to serious reduction of yield stress for the injection of the hydrogel and the capacity for loading therapeutic agents like indocyanine green (ICG) and doxorubicin (DOX) for the functions of photothermal therapy (PTT) and chemotherapy. The CDN hydrogel could thus be transported through a thin catheter and further in situ strengthened under physiological conditions, like in blood, by secondarily cross-linking with phosphate ions for longer degradation duration and better mechanical property. These characteristics met the requirements of arterial interventional embolization, which was demonstrated by renal embolism operation on rabbits, and meanwhile favored the inhibition of subcutaneous tumor growth on an animal model. Therefore, this work makes a breakthrough in the case of largely reducing the embolism risks, thus affording a novel generation for interventional embolization.
Collapse
Affiliation(s)
- Shili Ai
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Qinzong Gao
- Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Gele Cheng
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 101408, China
- Duke Kunshan University, Suzhou, Jiangsu 215316, China
| | - Pengfei Zhong
- Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Peiyu Cheng
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Yingying Ren
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Hao Wang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Xu Zhu
- Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Shanyue Guan
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaozhong Qu
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
31
|
Altun I, Demirlenk YM, Atar D, Cevik E, Gunduz S, Albadawi H, Oklu R. Advances and Challenges in Interventional Immuno-Oncology Locoregional Therapies. J Vasc Interv Radiol 2024; 35:164-172. [PMID: 38272636 DOI: 10.1016/j.jvir.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/15/2023] [Indexed: 01/27/2024] Open
Abstract
Interventional immuno-oncology is making strides in locoregional therapies to address complex tumor microenvironments. Long-standing interventional radiology cancer therapies, such as tumor ablation and embolization, are being recharacterized in the context of immunotherapy. Intratumoral injections, such as those of genetically engineered or unaltered viruses, and the delivery of immune cells, antibodies, proteins, or cytokines into targeted tumors, along with advancements in delivery techniques, have produced promising results in preliminary studies, indicating their antitumor effectiveness. Emerging strategies using DNA scaffolding, polysaccharides, glycan, chitosan, and natural products are also showing promise in targeted cancer therapy. The future of interventional immuno-oncology lies in personalized immunotherapies that capitalize on individual immune profiles and tumor characteristics, along with the exploration of combination therapies. This study will review various interventional immuno-oncology strategies and emerging technologies to enhance delivery of therapeutics and response to immunotherapy.
Collapse
Affiliation(s)
- Izzet Altun
- Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yusuf M Demirlenk
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona
| | - Dila Atar
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona
| | - Enes Cevik
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona
| | - Seyda Gunduz
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona; Department of Medical Oncology, Istinye University Bahcesehir Liv Hospital, Istanbul, Turkey
| | - Hassan Albadawi
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona
| | - Rahmi Oklu
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona.
| |
Collapse
|
32
|
de Barros NR, Gangrade A, Elsebahy A, Chen R, Zehtabi F, Ermis M, Falcone N, Haghniaz R, Khosravi S, Gomez A, Huang S, Mecwan M, Khorsandi D, Lee J, Zhu Y, Li B, Kim H, Thankam FG, Khademhosseini A. Injectable Nanoengineered Adhesive Hydrogel for Treating Enterocutaneous Fistulas. Acta Biomater 2024; 173:231-246. [PMID: 38465268 PMCID: PMC10919932 DOI: 10.1016/j.actbio.2023.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 03/12/2024]
Abstract
Enterocutaneous fistula (ECF) is a severe medical condition where an abnormal connection forms between the gastrointestinal tract and skin. ECFs are, in most cases, a result of surgical complications such as missed enterotomies or anastomotic leaks. The constant leakage of enteric and fecal contents from the fistula site leads to skin breakdown and increases the risk of infection. Despite advances in surgical techniques and postoperative management, ECF accounts for significant mortality rates, estimated between 15-20%, and causes debilitating morbidity. Therefore, there is a critical need for a simple and effective method to seal and heal ECF. Injectable hydrogels with combined properties of robust mechanical properties and cell infiltration/proliferation have the potential to block and heal ECF. Herein, we report the development of an injectable nanoengineered adhesive hydrogel (INAH) composed of a synthetic nanosilicate (Laponite®) and a gelatin-dopamine conjugate for treating ECF. The hydrogel undergoes fast cross-linking using a co-injection method, resulting in a matrix with improved mechanical and adhesive properties. INAH demonstrates appreciable blood clotting abilities and is cytocompatible with fibroblasts. The adhesive properties of the hydrogel are demonstrated in ex vivo adhesion models with skin and arteries, where the volume stability in the hydrated internal environment facilitates maintaining strong adhesion. In vivo assessments reveal that the INAH is biocompatible, supporting cell infiltration and extracellular matrix deposition while not forming fibrotic tissue. These findings suggest that this INAH holds promising translational potential for sealing and healing ECF.
Collapse
Affiliation(s)
- Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Ahmad Elsebahy
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - RunRun Chen
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Alejandro Gomez
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Shuyi Huang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Bingbing Li
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - HanJun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
- College of Pharmacy, Korea University, Sejong, Republic of Korea, 30019
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| |
Collapse
|
33
|
Zehtabi F, Gangrade A, Tseng K, Haghniaz R, Abasgholizadeh R, Montazerian H, Khorsandi D, Bahari J, Ahari A, Mohaghegh N, Kouchehbaghi NH, Mandal K, Mecwan M, Rashad A, de Barros NR, Byun Y, Ermis M, Kim HJ, Khademhosseini A. Injectable Shear-Thinning Hydrogels with Sclerosing and Matrix Metalloproteinase Modulatory Properties for the Treatment of Vascular Malformations. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2305880. [PMID: 38558868 PMCID: PMC10977963 DOI: 10.1002/adfm.202305880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Indexed: 04/04/2024]
Abstract
Sac embolization of abdominal aortic aneurysms (AAAs) remains clinically limited by endoleak recurrences. These recurrences are correlated with recanalization due to the presence of endothelial lining and matrix metalloproteinases (MMPs)-mediated aneurysm progression. This study incorporated doxycycline (DOX), a well-known sclerosant and MMPs inhibitor, into a shear-thinning biomaterial (STB)-based vascular embolizing hydrogel. The addition of DOX was expected to improve embolizing efficacy while preventing endoleaks by inhibiting MMP activity and promoting endothelial removal. The results showed that STBs containing 4.5% w/w silicate nanoplatelet and 0.3% w/v of DOX were injectable and had a 2-fold increase in storage modulus compared to those without DOX. STB-DOX hydrogels also reduced clotting time by 33% compared to untreated blood. The burst release of DOX from the hydrogels showed sclerosing effects after 6 h in an ex vivo pig aorta model. Sustained release of DOX from hydrogels on endothelial cells showed MMP inhibition (ca. an order of magnitude larger than control groups) after 7 days. The hydrogels successfully occluded a patient-derived abdominal aneurysm model at physiological blood pressures and flow rates. The sclerosing and MMP inhibition characteristics in the engineered multifunctional STB-DOX hydrogels may provide promising opportunities for the efficient embolization of aneurysms in blood vessels.
Collapse
Affiliation(s)
- Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Kaylee Tseng
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90007, United States
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Reza Abasgholizadeh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Hossein Montazerian
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Jamal Bahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Amir Ahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Negar Hosseinzadeh Kouchehbaghi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Textile Engineering, Amirkabir University of Technology (Tehran Polytechnic), Hafez Avenue, 1591634311 Tehran, Iran
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Ahmad Rashad
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | | | - Youngjoo Byun
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
- Vellore Institute of Technology (VIT), Vellore, India, 632014
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| |
Collapse
|
34
|
Shen W, Pei P, Zhang C, Li J, Han X, Liu T, Shi X, Su Z, Han G, Hu L, Yang K. A Polymeric Hydrogel to Eliminate Programmed Death-Ligand 1 for Enhanced Tumor Radio-Immunotherapy. ACS NANO 2023; 17:23998-24011. [PMID: 37988029 DOI: 10.1021/acsnano.3c08875] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Programmed death-ligand 1 (PD-L1) is a specialized shield on tumor cells that evades the immune system. Even inhibited by PD-L1 antibodies, a cycling process constantly transports PD-L1 from inside to outside of cells, facilitating the renewal and replenishment of PD-L1 on the cancer cell membrane. Herein, we develop a sodium alginate hydrogel consisting of elesclomol-Cu and galactose to induce persistent cuproptosis, leading to the reduction of PD-L1 for radio-immunotherapy of colon tumors. First, a prefabricated hydrogel is synthesized by immobilizing elesclomol onto a sodium alginate saccharide chain through the coordination with bivalent copper ions (Cu2+), followed by incorporation of galactose. After implantation into the tumors, this prefabricated hydrogel can be further cross-linked in the presence of physiological calcium ions (Ca2+), resulting in the formation of a hydrogel with controlled release of elesclomol-Cu2+ (ES-Cu) and galactose. The hydrogel effectively induces the oligomerization of DLAT and cuproptosis in colorectal cancer cells. Interestingly, radiation-induced PD-L1 upregulation is abrogated in the presence of the hydrogel, releasing ES-Cu and galactose. Consequently, the sensitization of tumor to radiotherapy and immunotherapy is significantly improved, further prolonging the survival of tumor-bearing mice in both local and metastatic tumors. Our study introduces an approach that combines cuproptosis with immunotherapy and radiotherapy.
Collapse
Affiliation(s)
- Wenhao Shen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, China
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Pei Pei
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Chonghai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, China
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Junmei Li
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215123, Jiangsu, China
| | - Xiangming Han
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215123, Jiangsu, China
| | - Teng Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, China
| | - Xiumin Shi
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, China
| | - Zhiyue Su
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215123, Jiangsu, China
| | - Gaohua Han
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Lin Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
35
|
Wang S, Qiu M, Liu J, Yin T, Wu C, Huang C, Han J, Cheng S, Peng Q, Li Y, Tie C, Wu X, Du S, Xu T. Preshaped 4D Photocurable Ultratough Organogel Microcoils for Personalized Endovascular Embolization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2308130. [PMID: 37962041 DOI: 10.1002/adma.202308130] [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: 08/11/2023] [Revised: 11/10/2023] [Indexed: 11/15/2023]
Abstract
Endovascular embolization using microcoils can be an effective technique to treat artery aneurysms. However, microcoils with fixed designs are difficult to adapt to all aneurysm types. In this paper, a photocurable ultratough shape memory organogel with a curing time of only 2 s and megapascal-level mechanical properties is proposed. Then, it is used to manufacture the personalized 4D microcoil with a wire diameter of only 0.3 mm. The improved mechanical modulus (511.63 MPa) can reduce the possibility of microcoils' fracture during embolization. Besides, the fast body-temperature-triggering shape memory ability makes the 4D microcoil applicable in vivo. These 4D microcoils are finally delivered into the rabbit, and successfully blocked the blood flow inside different aneurysms, with neoendothelial cells and collagen fibers growing on the microcoil surface snugly, indicating full aneurysm recovery. This 4D organogel microcoil can potentially be used in personalized clinical translation on human beings.
Collapse
Affiliation(s)
- Shu Wang
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| | - Ming Qiu
- Department of Neurosurgery, South China Hospital, Shenzhen University, Shenzhen, 518000, China
| | - Jiancheng Liu
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| | - Ting Yin
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan, 523000, China
| | - Chong Wu
- Department of Neurosurgery, South China Hospital, Shenzhen University, Shenzhen, 518000, China
| | - Chenyang Huang
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| | - Jianguo Han
- Department of Neurosurgery, South China Hospital, Shenzhen University, Shenzhen, 518000, China
| | - Si Cheng
- Department of Neurosurgery, South China Hospital, Shenzhen University, Shenzhen, 518000, China
| | - Qianbi Peng
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| | - Ye Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| | | | - Xinyu Wu
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| | - Shiwei Du
- Department of Neurosurgery, South China Hospital, Shenzhen University, Shenzhen, 518000, China
| | - Tiantian Xu
- Guangdong Provincial Key Lab of Robotics and Intelligent System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
- The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, China
| |
Collapse
|
36
|
Yang Y, Wang X, Yang F, Mu B, Wang A. Progress and future prospects of hemostatic materials based on nanostructured clay minerals. Biomater Sci 2023; 11:7469-7488. [PMID: 37873611 DOI: 10.1039/d3bm01326j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The occurrence of uncontrolled hemorrhage is a significant threat to human life and health. Although hemostatic materials have made remarkable advances in the biomaterials field, it remains a challenge to develop safe and effective hemostatic materials for global medical use. Natural clay minerals (CMs) have long been used as traditional inorganic hemostatic agents due to their good hemostatic capability, biocompatibility and easy availability. With the advancement of science, technology and ideology, CM-based hemostatic materials have undergone continuous innovations by integrating new inspirations with conventional concepts. This review systematically summarizes the hemostatic mechanisms of different natural CMs based on their nanostructures. Moreover, it also comprehensively reviews the latest research progress for CM-based hemostatic hybrid and nanocomposite materials, and discusses the challenges and developments in this field.
Collapse
Affiliation(s)
- Yinfeng Yang
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, P. R. China.
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, P. R. China
| | - Xiaomei Wang
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, P. R. China.
| | - Fangfang Yang
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, P. R. China.
| | - Bin Mu
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, P. R. China.
| | - Aiqin Wang
- Key Laboratory of Clay Mineral Applied Research of Gansu Province, Center of Eco-material and Green Chemistry, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, P. R. China.
| |
Collapse
|
37
|
Niu Y, Yang Z, Yang Y, Wang X, Zhang P, Lv L, Wang S, Liu Y, Liu Y, Zhou Y. Alkaline shear-thinning micro-nanocomposite hydrogels initiate endogenous TGFβ signaling for in situ bone regeneration. NPJ Regen Med 2023; 8:56. [PMID: 37833374 PMCID: PMC10575889 DOI: 10.1038/s41536-023-00333-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Recruiting endogenous stem cells to bone defects without stem cell transplantation and exogenous factor delivery represents a promising strategy for bone regeneration. Herein, we develop an alkaline shear-thinning micro-nanocomposite hydrogel (10-MmN), aiming to alkaline-activate endogenous TGFβ1 and achieve in situ bone regeneration. It contains polyethyleneimine (PEI)-modified gelatin, laponite nanoplatelets (LAP), a bicarbonate buffer with a pH of 10, and gelatin microspheres (MSs). PEI-modified gelatin plays a pivotal role in hydrogel fabrication. It endows the system with sufficient positive charges, and forms a shear-thinning nanocomposite matrix in the pH 10 buffer (10-mN) with negatively charged LAP via electrostatic gelation. For biological functions, the pH 10 buffer dominates alkaline activation of endogenous serum TGFβ1 to recruit rat bone marrow stem cells through the Smad pathway, followed by improved osteogenic differentiation. In addition, MSs are incorporated into 10-mN to form 10-MmN, and function as substrates to provide good attachment sites for the recruited stem cells and facilitate further their osteogenic differentiation. In a rat critical-sized calvarial defect model, 10-MmN exhibits excellent biocompatibility, biodegradability, hydrogel infusion and retention in bone defects with flexible shapes and active bleeding. Importantly, it repairs ~95% of the defect areas in 3 months by recruiting TGFβR2+ and CD90+CD146+ stem cells, and promoting cell proliferation, osteogenic differentiation and bone formation. The present study provides a biomaterial-based strategy to regulate alkalinity in bone defects for the initiation of endogenous TGFβ signaling, which can be extended to treat other diseases.
Collapse
Affiliation(s)
- Yuting Niu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| | - Zhen Yang
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yang Yang
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Xu Wang
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Ping Zhang
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Longwei Lv
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Sainan Wang
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
| | - Yunsong Liu
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
| | - Yongsheng Zhou
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Translational Research Center for Orocraniofacial Stem Cells and Systemic Health, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
| |
Collapse
|
38
|
Godau B, Samimi S, Seyfoori A, Samiei E, Khani T, Naserzadeh P, Najafabadi AH, Lesha E, Majidzadeh-A K, Ashtari B, Charest G, Morin C, Fortin D, Akbari M. A Drug-Eluting Injectable NanoGel for Localized Delivery of Anticancer Drugs to Solid Tumors. Pharmaceutics 2023; 15:2255. [PMID: 37765224 PMCID: PMC10534730 DOI: 10.3390/pharmaceutics15092255] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
Systemically administered chemotherapy reduces the efficiency of the anticancer agent at the target tumor tissue and results in distributed drug to non-target organs, inducing negative side effects commonly associated with chemotherapy and necessitating repeated administration. Injectable hydrogels present themselves as a potential platform for non-invasive local delivery vehicles that can serve as a slow-releasing drug depot that fills tumor vasculature, tissue, or resection cavities. Herein, we have systematically formulated and tested an injectable shear-thinning hydrogel (STH) with a highly manipulable release profile for delivering doxorubicin, a common chemotherapeutic. By detailed characterization of the STH physical properties and degradation and release dynamics, we selected top candidates for testing in cancer models of increasing biomimicry. Two-dimensional cell culture, tumor-on-a-chip, and small animal models were used to demonstrate the high anticancer potential and reduced systemic toxicity of the STH that exhibits long-term (up to 80 days) doxorubicin release profiles for treatment of breast cancer and glioblastoma. The drug-loaded STH injected into tumor tissue was shown to increase overall survival in breast tumor- and glioblastoma-bearing animal models by 50% for 22 days and 25% for 52 days, respectively, showing high potential for localized, less frequent treatment of oncologic disease with reduced dosage requirements.
Collapse
Affiliation(s)
- Brent Godau
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Sadaf Samimi
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Amir Seyfoori
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ehsan Samiei
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Tahereh Khani
- Preclinical Lab., Core Facility, Tehran University of Medical Sciences, Tehran 1417755354, Iran
| | - Parvaneh Naserzadeh
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran 88945173, Iran
| | | | - Emal Lesha
- Department of Neurosurgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, No. 146, South Gandhi Ave., Vanak Sq., P.O. BOX 1517964311, Tehran 1684613114, Iran
| | - Behnaz Ashtari
- Department of Medical Nanotechnology, Faculty of Advance Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Gabriel Charest
- Department of Surgery, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada (C.M.); (D.F.)
| | - Christophe Morin
- Department of Surgery, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada (C.M.); (D.F.)
| | - David Fortin
- Department of Surgery, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada (C.M.); (D.F.)
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
- Terasaki Institute for Biomedical Innovations, Los Angeles, CA 90050, USA;
| |
Collapse
|
39
|
de Barros NR, Gomez A, Ermis M, Falcone N, Haghniaz R, Young P, Gao Y, Aquino AF, Li S, Niu S, Chen R, Huang S, Zhu Y, Eliahoo P, Sun A, Khorsandi D, Kim J, Kelber J, Khademhosseini A, Kim HJ, Li B. Gelatin methacryloyl and Laponite bioink for 3D bioprinted organotypic tumor modeling. Biofabrication 2023; 15:10.1088/1758-5090/ace0db. [PMID: 37348491 PMCID: PMC10683563 DOI: 10.1088/1758-5090/ace0db] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/22/2023] [Indexed: 06/24/2023]
Abstract
Three-dimensional (3D)in vitrotumor models that can capture the pathophysiology of human tumors are essential for cancer biology and drug development. However, simulating the tumor microenvironment is still challenging because it consists of a heterogeneous mixture of various cellular components and biological factors. In this regard, current extracellular matrix (ECM)-mimicking hydrogels used in tumor tissue engineering lack physical interactions that can keep biological factors released by encapsulated cells within the hydrogel and improve paracrine interactions. Here, we developed a nanoengineered ion-covalent cross-linkable bioink to construct 3D bioprinted organotypic tumor models. The bioink was designed to implement the tumor ECM by creating an interpenetrating network composed of gelatin methacryloyl (GelMA), a light cross-linkable polymer, and synthetic nanosilicate (Laponite) that exhibits a unique ionic charge to improve retention of biological factors released by the encapsulated cells and assist in paracrine signals. The physical properties related to printability were evaluated to analyze the effect of Laponite hydrogel on bioink. Low GelMA (5%) with high Laponite (2.5%-3.5%) composite hydrogels and high GelMA (10%) with low Laponite (1.0%-2.0%) composite hydrogels showed acceptable mechanical properties for 3D printing. However, a low GelMA composite hydrogel with a high Laponite content could not provide acceptable cell viability. Fluorescent cell labeling studies showed that as the proportion of Laponite increased, the cells became more aggregated to form larger 3D tumor structures. Reverse transcription-polymerase chain reaction (RT-qPCR) and western blot experiments showed that an increase in the Laponite ratio induces upregulation of growth factor and tissue remodeling-related genes and proteins in tumor cells. In contrast, cell cycle and proliferation-related genes were downregulated. On the other hand, concerning fibroblasts, the increase in the Laponite ratio indicated an overall upregulation of the mesenchymal phenotype-related genes and proteins. Our study may provide a rationale for using Laponite-based hydrogels in 3D cancer modeling.
Collapse
Affiliation(s)
- Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Alejandro Gomez
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
- Department of Biology, California State University, Northridge, CA 91330, United States of America
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- METU Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Patric Young
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Yaqi Gao
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
| | - Albert-Fred Aquino
- Department of Biology, California State University, Northridge, CA 91330, United States of America
| | - Siyuan Li
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
- Department of Biomedical Engineering, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, United States of America
| | - Siyi Niu
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
- Department of Biology, University of California, Irvine, CA 92697, United States of America
| | - RunRun Chen
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
| | - Shuyi Huang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Payam Eliahoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90088, United States of America
| | - Arthur Sun
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
- Department of Integrative Biology, University of California, Berkeley, CA 94720, United States of America
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Jinjoo Kim
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Jonathan Kelber
- Department of Biology, California State University, Northridge, CA 91330, United States of America
- Department of Biology, Baylor University, 101 Bagby Ave, TX 76706, United States of America
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Bingbing Li
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
| |
Collapse
|
40
|
Jin D, Wang Q, Chan KF, Xia N, Yang H, Wang Q, Yu SCH, Zhang L. Swarming self-adhesive microgels enabled aneurysm on-demand embolization in physiological blood flow. SCIENCE ADVANCES 2023; 9:eadf9278. [PMID: 37172097 PMCID: PMC10181194 DOI: 10.1126/sciadv.adf9278] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The recent rise of swarming microrobotics offers great promise in the revolution of minimally invasive embolization procedure for treating aneurysm. However, targeted embolization treatment of aneurysm using microrobots has significant challenges in the delivery capability and filling controllability. Here, we develop an interventional catheterization-integrated swarming microrobotic platform for aneurysm on-demand embolization in physiological blood flow. A pH-responsive self-healing hydrogel doped with magnetic and imaging agents is developed as the embolic microgels, which enables long-term self-adhesion under biological condition in a controllable manner. The embolization strategy is initiated by catheter-assisted deployment of swarming microgels, followed by the application of external magnetic field for targeted aggregation of microrobots into aneurysm sac under the real-time guidance of ultrasound and fluoroscopy imaging. Mild acidic stimulus is applied to trigger the welding of microgels with satisfactory bio-/hemocompatibility and physical stability and realize complete embolization. Our work presents a promising connection between the design and control of microrobotic swarms toward practical applications in dynamic environments.
Collapse
Affiliation(s)
- Dongdong Jin
- School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen, 518071, Guangdong, China
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
| | - Qinglong Wang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
| | - Kai Fung Chan
- Chow Yuk Ho Technology Centre for Innovative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
| | - Neng Xia
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
| | - Haojin Yang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
| | - Qianqian Wang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing 211000, China
| | - Simon Chun Ho Yu
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
- Vascular and Interventional Radiology Foundation Clinical Science Centre, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
| | - Li Zhang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
- Chow Yuk Ho Technology Centre for Innovative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
- T-Stone Robotics Institute, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 999077, China
| |
Collapse
|
41
|
Pal A, Blanzy J, Gómez KJR, Preul MC, Vernon BL. Liquid Embolic Agents for Endovascular Embolization: A Review. Gels 2023; 9:gels9050378. [PMID: 37232970 DOI: 10.3390/gels9050378] [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: 03/06/2023] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 05/27/2023] Open
Abstract
Endovascular embolization (EE) has been used for the treatment of blood vessel abnormalities, including aneurysms, AVMs, tumors, etc. The aim of this process is to occlude the affected vessel using biocompatible embolic agents. Two types of embolic agents, solid and liquid, are used for endovascular embolization. Liquid embolic agents are usually injectable and delivered into the vascular malformation sites using a catheter guided by X-ray imaging (i.e., angiography). After injection, the liquid embolic agent transforms into a solid implant in situ based on a variety of mechanisms, including polymerization, precipitation, and cross-linking, through ionic or thermal process. Until now, several polymers have been designed successfully for the development of liquid embolic agents. Both natural and synthetic polymers have been used for this purpose. In this review, we discuss embolization procedures with liquid embolic agents in different clinical applications, as well as in pre-clinical research studies.
Collapse
Affiliation(s)
- Amrita Pal
- Center for Interventional Biomaterials, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Jeffrey Blanzy
- Center for Interventional Biomaterials, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Karime Jocelyn Rosas Gómez
- Center for Interventional Biomaterials, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| | - Mark C Preul
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Brent L Vernon
- Center for Interventional Biomaterials, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
42
|
Dobashi Y, Ku JC, Ramjist J, Pasarikovski C, Walus K, Madden JDW, Yang VXD. Photomodulated Extrusion as a Localized Endovascular Hydrogel Deposition Method. Adv Healthc Mater 2023; 12:e2202632. [PMID: 36681868 PMCID: PMC11468792 DOI: 10.1002/adhm.202202632] [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: 10/12/2022] [Revised: 01/10/2023] [Indexed: 01/23/2023]
Abstract
Minimally invasive endovascular embolization is used to treat a wide range of diseases in neurology, oncology, and trauma where the vascular morphologies and corresponding hemodynamics vary greatly. Current techniques based on metallic coils, flow diverters, liquid embolics, and suspended microspheres are limited in their ability to address a wide variety of vasculature and can be plagued by complications including distal migration, compaction, and inappropriate vascular remodeling. Further, these endovascular devices currently offer limited therapeutic functions beyond flow control such as drug delivery. Herein, a novel in situ microcatheter-based photomodulated extrusion approach capable of dynamically tuning the physical and morphological properties of injectable hydrogels, optimizing for local hemodynamic environment and vascular morphology, is proposed and demonstrated. A shear thinning and photoactivated poly(ethylene glycol diacrylate)-nanosilicate (PEGDA-nSi) hydrogel is used to demonstrate multiple extrusion modes which are controlled by photokinetics and device configurations. Real-time photomodulation of injected hydrogel viscosity and modulus is successfully used for embolization in various vasculatures, including high-flow large vessels and arterial-to-arterial capillary shunts. Furthermore, a generalizable therapeutic delivery platform is proposed by demonstrating a core-shell structured extrusion encapsulating doxorubicin to achieve a more sustained release compared to unencapsulated payload.
Collapse
Affiliation(s)
- Yuta Dobashi
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioM5S 1A1Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Jerry C. Ku
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioM5S 1A1Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
- Division of NeurosurgeryDepartment of SurgeryUniversity of TorontoTorontoOntarioM5S 1A1Canada
| | - Joel Ramjist
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Christopher Pasarikovski
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioM5S 1A1Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
- Division of NeurosurgeryDepartment of SurgeryUniversity of TorontoTorontoOntarioM5S 1A1Canada
| | - Konrad Walus
- Department of Electrical and Computer EngineeringSchool of Biomedical EngineeringUniversity of British ColumbiaVancouverBritish ColumbiaV6T 1Z4Canada
| | - John D. W. Madden
- Department of Electrical and Computer EngineeringSchool of Biomedical EngineeringUniversity of British ColumbiaVancouverBritish ColumbiaV6T 1Z4Canada
| | - Victor X. D. Yang
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioM5S 1A1Canada
- Division of NeurosurgeryDepartment of SurgeryUniversity of TorontoTorontoOntarioM5S 1A1Canada
| |
Collapse
|
43
|
Haghniaz R, Kim HJ, Montazerian H, Baidya A, Tavafoghi M, Chen Y, Zhu Y, Karamikamkar S, Sheikhi A, Khademhosseini A. Tissue adhesive hemostatic microneedle arrays for rapid hemorrhage treatment. Bioact Mater 2023; 23:314-327. [PMID: 36439081 PMCID: PMC9692134 DOI: 10.1016/j.bioactmat.2022.08.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/06/2022] [Accepted: 08/20/2022] [Indexed: 11/26/2022] Open
Abstract
Blood loss by hemorrhaging wounds accounts for over one-third of ∼5 million trauma fatalities worldwide every year. If not controlled in a timely manner, exsanguination can take lives within a few minutes. Developing new biomaterials that are easy to use by non-expert patients and promote rapid blood coagulation is an unmet medical need. Here, biocompatible, and biodegradable microneedle arrays (MNAs) based on gelatin methacryloyl (GelMA) biomaterial hybridized with silicate nanoplatelets (SNs) are developed for hemorrhage control. The SNs render the MNAs hemostatic, while the needle-shaped structure increases the contact area with blood, synergistically accelerating the clotting time from 11.5 min to 1.3 min in vitro. The engineered MNAs reduce bleeding by ∼92% compared with the untreated injury group in a rat liver bleeding model. SN-containing MNAs outperform the hemostatic effect of needle-free patches and a commercial hemostat in vivo via combining micro- and nanoengineered features. Furthermore, the tissue adhesive properties and mechanical interlocking support the suitability of MNAs for wound closure applications. These hemostatic MNAs may enable rapid hemorrhage control, particularly for patients in developing countries or remote areas with limited or no immediate access to hospitals.
Collapse
Affiliation(s)
- Reihaneh Haghniaz
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, CA, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, CA, 90024, United States
| | - Han-Jun Kim
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, CA, 90024, United States
| | - Hossein Montazerian
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, CA, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, CA, 90024, United States
| | - Avijit Baidya
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Maryam Tavafoghi
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, CA, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Yi Chen
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, CA, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, CA, 90024, United States
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, CA, 90024, United States
| | - Solmaz Karamikamkar
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, CA, 90024, United States
| | - Amir Sheikhi
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, CA, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, United States
| | - Ali Khademhosseini
- Department of Bioengineering, University of California, Los Angeles, 410 Westwood Plaza, Los Angeles, CA, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA, 90095, United States
- Terasaki Institute for Biomedical Innovation, 11570 W Olympic Boulevard, Los Angeles, CA, 90024, United States
| |
Collapse
|
44
|
Mecwan M, Haghniaz R, Najafabadi AH, Mandal K, Jucaud V, John JV, Khademhosseini A. Thermoresponsive shear-thinning hydrogel (T-STH) hemostats for minimally invasive treatment of external hemorrhages. Biomater Sci 2023; 11:949-963. [PMID: 36537259 DOI: 10.1039/d2bm01559e] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hemorrhage is the leading cause of death following battlefield injuries. Although several hemostats are commercially available, they do not meet all the necessary requirements to stop bleeding in combat injuries. Here, we engineer thermoresponsive shear-thinning hydrogels (T-STH) composed of a thermoresponsive polymer, poly(N-isopropyl acrylamide) (p(NIPAM)), and hemostatic silicate nanodisks, LAPONITE®, as minimally invasive injectable hemostatic agents. Our T-STH is a physiologically stable hydrogel that can be easily injected through a syringe and needle and exhibits rapid mechanical recovery. Additionally, it demonstrates temperature-dependent blood coagulation owing to the phase transition of p(NIPAM). It decreases in vitro blood clotting times over 50% at physiological temperatures compared to room temperature. Furthermore, it significantly prevents blood loss in an ex vivo bleeding model at different blood flow rates (1 mL min-1 and 5 mL min-1) by forming a wound plug. More importantly, our T-STH is comparable to a commercially available hemostat, Floseal, in terms of blood loss and blood clotting time in an in vivo rat liver bleeding model. Furthermore, once the hemorrhage is stabilized, our T-STH can be easily removed using a cold saline wash without any rebleeding or leaving any residues. Taken together, our T-STH can be used as a first aid hemostat to treat external hemorrhages in emergency situations.
Collapse
Affiliation(s)
- Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA.
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA.
| | | | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA.
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA.
| | - Johnson V John
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA.
| |
Collapse
|
45
|
Stewart RJ, Sima M, Karz J, Jones JP. Material characterization of GPX ®: A versatile in situ solidifying embolic platform technology. Front Bioeng Biotechnol 2023; 11:1095148. [PMID: 36726745 PMCID: PMC9885798 DOI: 10.3389/fbioe.2023.1095148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
Endovascular embolization is a minimally invasive procedure during which blood flow to targeted tissues is selectively occluded. The list of clinical indications for embolization continues to expand. Liquid embolic agents are injectable compositions that transition into a solid or semi-solid form when introduced into blood vessels. The mechanism that triggers the liquid-to-solid transition is a key distinguishing feature of liquid embolic agents. GPX is a waterborne liquid embolic agent comprising oppositely charged polyelectrolytes: polyguanidinum and inorganic polyphoshate. In situ solidification is driven by electrostatic condensation of the polyelectrolytes, triggered by ionic strength differentials. We report in vitro characterization of the material properties of GPX, it is in vivo effectiveness in acute animal studies, and its potential for chemoembolization. The viscosity of GPX can be varied over a wide range by adjusting the polyguanidinium MW and/or concentration. Formulation of GPX with either tantalum microparticles (30 wt%) or iodinated radiocontrast agents (300 mgI ml-1) did not significantly change the flow behavior of GPX; the viscosity was independent of shear rate and remained within a clinically practical range (80-160 cP). Formulation of GPX with doxorubicin substantially increased viscosity at low shear rates and resulted in a power law dependence on shear rate. High contrast and effective vascular occlusion were demonstrated in both swine kidneys and rete mirabile. Contrast from iodinated compounds was temporary, dissipating within hours. The doxorubicin in vitro release profile was linear over 90 days. The results demonstrate that GPX is a versatile liquid embolic platform that can be formulated with a wide range of viscosities injectable at clinically practical flow rates, with either transient or permanent contrast, and that can provide prolonged zero-order delivery of doxorubicin to embolized tissues.
Collapse
Affiliation(s)
- Russell J. Stewart
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States,*Correspondence: Russell J. Stewart,
| | - Monika Sima
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Jessica Karz
- Fluidx Medical Technology, Inc., Salt Lake City, UT, United States
| | - Joshua P. Jones
- Fluidx Medical Technology, Inc., Salt Lake City, UT, United States
| |
Collapse
|
46
|
Chen S, Zhao R, Sun X, Wang H, Li L, Liu J. Toxicity and Biocompatibility of Liquid Metals. Adv Healthc Mater 2023; 12:e2201924. [PMID: 36314401 DOI: 10.1002/adhm.202201924] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/15/2022] [Indexed: 01/27/2023]
Abstract
Recently, room-temperature liquid metals have attracted increasing attention from researchers owing to their excellent material properties. Systematic interpretation of the potential toxicity issues involved is essential for a wide range of applications, especially in the biomedical and healthcare fields. However, even with the exponential growth of related studies, investigation of the toxicological impact and possible hazards of liquid metals to organisms is still in its infancy. This review aims to provide a comprehensive summary of the current frontier of knowledge on liquid metal toxicology and biocompatibility in different environments. Based on recent studies, this review focuses on Ga and Bi-based in different states. It is necessary to evaluate their toxicity considering the rapid increase in research and utilization of such liquid metal composites. Finally, existing challenges are discussed and suggestions are provided for further investigation of liquid metal toxicology to clarify the toxicological mechanisms and strategies are provided to avoid adverse effects. In addition to resolving the doubts of public concern about the toxicity of liquid metals, this review is expected to promote the healthy and sustainable development of liquid metal-based materials and their use in diverse areas, especially those related to health care.
Collapse
Affiliation(s)
- Sen Chen
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ruiqi Zhao
- Beijing Key Lab of Cryo-Biomedical Engineering and Key Lab of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xuyang Sun
- School of Medicine Engineering, Beijing University of Aeronautics and Astronautics, Beijing, 100191, China
| | - Hongzhang Wang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Lei Li
- Beijing Key Lab of Cryo-Biomedical Engineering and Key Lab of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jing Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China.,Beijing Key Lab of Cryo-Biomedical Engineering and Key Lab of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
47
|
Zehtabi F, Montazerian H, Haghniaz R, Tseng K, Mohaghegh N, Mandal K, Zamanian B, Dokmeci MR, Akbari M, Najafabadi AH, Kim HJ, Khademhosseini A. Sodium Phytate-Incorporated Gelatin-Silicate Nanoplatelet Composites for Enhanced Cohesion and Hemostatic Function of Shear-Thinning Biomaterials. Macromol Biosci 2023; 23:e2200333. [PMID: 36287084 PMCID: PMC9851971 DOI: 10.1002/mabi.202200333] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/22/2022] [Indexed: 01/22/2023]
Abstract
Shear-thinning biomaterials (STBs) based on gelatin-silicate nanoplatelets (SNs) are emerging as an alternative to conventional coiling and clipping techniques in the treatment of vascular anomalies. Improvements in the cohesion of STB hydrogels pave the way toward their translational application in minimally invasive therapies such as endovascular embolization repair. In the present study, sodium phytate (Phyt) additives are used to tune the electrostatic network of SNs-gelatin STBs, thereby promoting their mechanical integrity and facilitating injectability through standard catheters. We show that an optimized amount of Phyt enhances storage modulus by approximately one order of magnitude and reduces injection force by ≈58% without compromising biocompatibility and hydrogel wet stability. The Phyt additives are found to decrease the immune responses induced by SNs. In vitro embolization experiments suggest a significantly lower rate of failure in Phyt-incorporated STBs than in control groups. Furthermore, the addition of Phyt leads to accelerated blood coagulation (reduces clotting time by ≈45% compared to controls) due to the contributions of negatively charged phosphate groups, which aid in the prolonged durability of STB in coagulopathic patients. Therefore, the proposed approach is an effective method for the design of robust and injectable STBs for minimally invasive treatment of vascular malformations.
Collapse
Affiliation(s)
- Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Hossein Montazerian
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Kaylee Tseng
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90007, United States
| | - Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Behnam Zamanian
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Mohsen Akbari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
- Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100, Gliwice, Poland
| | | | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| |
Collapse
|
48
|
Song M, Wang J, He J, Kan D, Chen K, Lu J. Synthesis of Hydrogels and Their Progress in Environmental Remediation and Antimicrobial Application. Gels 2022; 9:16. [PMID: 36661783 PMCID: PMC9858390 DOI: 10.3390/gels9010016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
As a kind of efficient adsorptive material, hydrogel has a wide application prospect within different fields, owing to its unique 3D network structures composed of polymers. In this paper, different synthetic strategies, crosslinking methods and their corresponding limitations and outstanding contributions of applications in the fields of removing environmental pollutants are reviewed to further provide a prospective view of their applications in water resources sustainability. Furthermore, the applications within the biomedical field, especially in wound dressing, are also reviewed in this paper, mainly due to their unique water retention ability, antibacterial ability, and good biocompatibility. Finally, the development direction of hydrogels in the fields of environmental remediation and biomedicine were summarized and prospected.
Collapse
Affiliation(s)
- Mengshan Song
- Advanced Materials Research Central, Northwest Institute for Nonferrous Metal Research, Xi’an 710016, China
| | - Jingfeng Wang
- Advanced Materials Research Central, Northwest Institute for Nonferrous Metal Research, Xi’an 710016, China
| | - Jiabei He
- Advanced Materials Research Central, Northwest Institute for Nonferrous Metal Research, Xi’an 710016, China
| | - Dongxiao Kan
- Advanced Materials Research Central, Northwest Institute for Nonferrous Metal Research, Xi’an 710016, China
| | - Kaiyun Chen
- Advanced Materials Research Central, Northwest Institute for Nonferrous Metal Research, Xi’an 710016, China
| | - Jialu Lu
- Shanghai Key Laboratory of Special Artificial Microstructure Materials and Technology, School of Physics Science and Engineering, Tongji University, Shanghai 200092, China
| |
Collapse
|
49
|
Ku JC, Dobashi Y, Pasarikovski CR, Ramjist J, Hamani C, Heyn C, Walus K, Yang VXD. Photosensitive Hydrogel-Based Embolic Agent Treatment of Wide-Necked Aneurysms: Preliminary Animal Results. Gels 2022; 8:gels8120788. [PMID: 36547312 PMCID: PMC9778217 DOI: 10.3390/gels8120788] [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: 10/01/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Background: The endovascular treatment of cerebral aneurysms has become widespread but may still be limited by recurrence rates or complications. The discovery of novel embolic strategies may help mitigate these concerns. Methods: We formulated a Photosensitive Hydrogel Polymer (PHP) embolic agent which is low-viscosity, shear-thinning, and radio-opaque. After the filling of an aneurysm with PHP with balloon assistance, we utilized photopolymerization to induce solidification. Different methods of light delivery for photopolymerization were assessed in silicone models of aneurysms and in four acute animal trials with venous anastomosis aneurysms in pigs. Then, balloon-assisted embolization with PHP and photopolymerization was performed in three aneurysms in pigs with a one-month follow-up. Filling volume, recurrence rates, and complications were recorded. Results: The PHP was found to be suitable for the intravascular delivery and treatment of cerebral aneurysms. It was found that light delivery through the balloon catheter, as opposed to light delivery through the injection microcatheter, led to higher rates of filling in the 3D model and acute animal model for cerebral aneurysms. Using the balloon-assisted embolization and light delivery strategy, three wide-necked aneurysms were treated without complication. One-month follow-up showed no recurrence or neck remnants. Conclusions: We demonstrated a novel method of balloon-assisted photosensitive hydrogel polymer embolization and photopolymerization, leading to complete aneurysm filling with no recurrence at 1 month in three wide-necked aneurysms in pigs. This promising methodology will be investigated further with longer-term comparative animal trials.
Collapse
Affiliation(s)
- Jerry C. Ku
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence:
| | - Yuta Dobashi
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Joel Ramjist
- Department of Electrical, Computer and Biomedical Engineering, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | - Clement Hamani
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chinthaka Heyn
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Neuroradiology, Department of Radiology, University of Toronto, Toronto, ON M5T 1W7, Canada
| | - Konrad Walus
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Victor X. D. Yang
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Electrical, Computer and Biomedical Engineering, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
- Division of Neurosurgery, Department of Clinical Neurological Sciences, Schulich School of Medicine, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
50
|
Piezoelectric MoS2 Nanoflowers (NF's) for Targeted Cancer Therapy by Gelatin-based Shear Thinning Hydrogels. In vitro and In vivo trials. REACT FUNCT POLYM 2022. [DOI: 10.1016/j.reactfunctpolym.2022.105435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|