1
|
Kra JA, Markosian C, Tang FHF, de León AB, Chundury A, Agarwalla PK, Staquicini DI, Pasqualini R, Arap W. Brain-derived textiloma post glioblastoma resection and application of oxidized regenerated cellulose: A pilot, bedside-to-bench, translational study. Brain Pathol 2025; 35:e13331. [PMID: 39838550 PMCID: PMC12145898 DOI: 10.1111/bpa.13331] [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: 08/02/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
Oxidized regenerated cellulose (ORC; marketed as Surgicel® and Tabotamp®) is routinely used as an intraoperative hemostatic agent. Rarely, residual ORC has been associated with a foreign body reaction generating cystic or granulomatous lesions (i.e., textilomas) at the surgical site. Here, we report a bedside-to-bench, translational report of an intracranial mass after neurosurgical resection of glioblastoma with ORC application. As part of patient care, we performed magnetic resonance imaging and histopathological analysis of the mass. We then performed in vitro studies to evaluate the effect of ORC on cytokine production and viability of BV-2 murine microglial cells by using quantitative PCR along with live cell microscopy and crystal violet staining, respectively. Magnetic resonance imaging demonstrated a recurrent mass pressing on the adjacent right ventricle, which was removed in a second surgery for diagnostic and therapeutic purposes. Unexpectedly, histopathological examination of the resected mass revealed abundant ORC arising from the site with inflammation, microglial activation, and collagenization. Mechanistically, we show an ORC-induced modest increase in inflammatory cytokines with a subsequent decrease in microglial cell viability. These findings suggest that ORC may mediate microglial immune response and viability, and serve to raise awareness and guide interpretation of post-treatment surveillance imaging findings in the instance of foreign body reaction.
Collapse
Affiliation(s)
- Joshua A. Kra
- Rutgers Cancer InstituteNewarkNew JerseyUSA
- Division of Hematology/Oncology, Department of MedicineRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Christopher Markosian
- Rutgers Cancer InstituteNewarkNew JerseyUSA
- Division of Cancer Biology, Department of Radiation OncologyRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Fenny H. F. Tang
- Rutgers Cancer InstituteNewarkNew JerseyUSA
- Division of Cancer Biology, Department of Radiation OncologyRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Ada Baisre de León
- Department of Pathology, Immunology, and Laboratory MedicineRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Anupama Chundury
- Rutgers Cancer InstituteNew BrunswickNew JerseyUSA
- Department of Radiation OncologyRutgers Robert Wood Johnson Medical SchoolNew BrunswickNew JerseyUSA
- Present address:
Department of Radiation Oncology, Stritch School of MedicineLoyola University ChicagoMaywoodIllinoisUSA
| | - Pankaj K. Agarwalla
- Rutgers Cancer InstituteNewarkNew JerseyUSA
- Department of Neurological SurgeryRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Daniela I. Staquicini
- Rutgers Cancer InstituteNewarkNew JerseyUSA
- Division of Cancer Biology, Department of Radiation OncologyRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Renata Pasqualini
- Rutgers Cancer InstituteNewarkNew JerseyUSA
- Division of Cancer Biology, Department of Radiation OncologyRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| | - Wadih Arap
- Rutgers Cancer InstituteNewarkNew JerseyUSA
- Division of Hematology/Oncology, Department of MedicineRutgers New Jersey Medical SchoolNewarkNew JerseyUSA
| |
Collapse
|
2
|
Tan X, Xi H, Xue P, Cao J, Yarmolenko MA, Liu X, Jiang X. The gelatin sponge loaded with curcumin coating exhibits a synergistic effect of hemostasis, anti-inflammatory, and anti-scarring. BIOMATERIALS ADVANCES 2025; 169:214155. [PMID: 39709690 DOI: 10.1016/j.bioadv.2024.214155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
Postoperative wound healing has been extensively studied and well-documented. Gelatin sponges are commonly used in surgeries for blood absorption. If these sponges can also release drugs with anti-scarring and anti-inflammatory effects, they would significantly enhance wound healing. In this study, we investigated for the first time the application of curcumin films on the surface of gelatin sponges with high hemostatic efficiency using the Electron Beam Deposition (EBD) method. The structure of curcumin was analyzed using 1H NMR, FT-IR and XPS techniques. We examined the influence of the film on the sponge's absorption capacity and the impact of the sponge on drug release kinetics. Results showed that the presence of the curcumin film did not compromise the sponge's hemostatic ability. Additionally, compared to a flat substrate, the curcumin film on this highly porous substrate facilitated better curcumin release. Further experiments, including cytotoxicity tests, live/dead double staining, western blotting, and a scar model in mice, demonstrated that gelatin sponges with curcumin films exhibit a synergistic effect, combining anti-scarring, anti-inflammatory, and hemostatic properties.
Collapse
Affiliation(s)
- Xiaoxue Tan
- International Chinese-Belorussian Scientific laboratory on Vacuum-Plasma Technology, Nanjing University of Science and Technology, 200, Xiaolingwei street, Nanjing 210094, China
| | - Hongzhong Xi
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Orthopedics, Nanjing 210029, China
| | - Peng Xue
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Orthopedics, Nanjing 210029, China
| | - Jinxin Cao
- Nanjing Xiaozhuang University, Nanjing 211171, China
| | - M A Yarmolenko
- International Chinese-Belorussian Scientific laboratory on Vacuum-Plasma Technology, Nanjing University of Science and Technology, 200, Xiaolingwei street, Nanjing 210094, China; Francisk Skorina Gomel State University, 104, Sovetskaya street, Gomel 246019, Belarus
| | - Xin Liu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Orthopedics, Nanjing 210029, China.
| | - Xiaohong Jiang
- International Chinese-Belorussian Scientific laboratory on Vacuum-Plasma Technology, Nanjing University of Science and Technology, 200, Xiaolingwei street, Nanjing 210094, China.
| |
Collapse
|
3
|
Wang Y, Che H, Qu L, Lu X, Dong M, Sun B, Guan H. The role of nanomaterials in revolutionizing ischemic stroke treatment: Current trends and future prospects. iScience 2024; 27:111373. [PMID: 39669428 PMCID: PMC11634991 DOI: 10.1016/j.isci.2024.111373] [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] [Indexed: 12/14/2024] Open
Abstract
Ischemic stroke has a high disability rate, which leads to irreversible neuronal death. The efficacy of conventional stroke treatments, including thrombolytic and neuroprotective therapies, is constrained by a number of factors, including safety concerns and inefficient drug delivery. The advent of nanomaterials has created new avenues for stroke therapy, facilitating enhanced pharmacokinetic behavior of drugs, effective drug accumulation at the target site, augmented therapeutic efficacy, and concomitant reduction in side effects. Therefore, this paper pioneers a research approach that summarized the development trend and clinical value of nanomaterials in the field of ischemic stroke through bibliometric analysis. This review provides an overview of the pathophysiological mechanisms of stroke and examines the current research trends in the use of nanomaterials in stroke management. It encompasses a multitude of domains, including targeted drug delivery systems, biosensors for the sensitive detection of biomarkers, and neuroprotective nanotechnologies capable of traversing the blood-brain barrier. Moreover, we investigate the challenges that nanomaterials encounter in the clinical translation context, including those pertaining to biocompatibility and long-term safety. These results have provided the clinical value and limitations of nanomaterials in the diagnosis and treatment of ischemic stroke from double perspectives, thereby offering new avenues for the further development of innovative nanotherapeutic tools.
Collapse
Affiliation(s)
- Yong Wang
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Huiying Che
- Department of General Practice, Yanbian University Hospital, Yanji 133002, China
| | - Linzhuo Qu
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Xin Lu
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Mingzhen Dong
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Bo Sun
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| | - Hongjian Guan
- Stroke Center, Department of Neurology, Yanbian University Hospital, Yanji 133002, China
| |
Collapse
|
4
|
Rotaru-Zăvăleanu AD, Dinescu VC, Aldea M, Gresita A. Hydrogel-Based Therapies for Ischemic and Hemorrhagic Stroke: A Comprehensive Review. Gels 2024; 10:476. [PMID: 39057499 PMCID: PMC11276304 DOI: 10.3390/gels10070476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Stroke remains the second leading cause of death and a major cause of disability worldwide, significantly impacting individuals, families, and healthcare systems. This neurological emergency can be triggered by ischemic events, including small vessel arteriolosclerosis, cardioembolism, and large artery atherothromboembolism, as well as hemorrhagic incidents resulting from macrovascular lesions, venous sinus thrombosis, or vascular malformations, leading to significant neuronal damage. The resultant motor impairment, cognitive dysfunction, and emotional disturbances underscore the urgent need for effective therapeutic interventions. Recent advancements in biomaterials, particularly hydrogels, offer promising new avenues for stroke management. Hydrogels, composed of three-dimensional networks of hydrophilic polymers, are notable for their ability to absorb and retain substantial amounts of water. Commonly used polymers in hydrogel formulations include natural polymers like alginate, chitosan, and collagen, as well as synthetic polymers such as polyethylene glycol (PEG), polyvinyl alcohol (PVA), and polyacrylamide. Their customizable characteristics-such as their porosity, swelling behavior, mechanical strength, and degradation rates-make hydrogels ideal for biomedical applications, including drug delivery, cell delivery, tissue engineering, and the controlled release of therapeutic agents. This review comprehensively explores hydrogel-based approaches to both ischemic and hemorrhagic stroke therapy, elucidating the mechanisms by which hydrogels provide neuroprotection. It covers their application in drug delivery systems, their role in reducing inflammation and secondary injury, and their potential to support neurogenesis and angiogenesis. It also discusses current advancements in hydrogel technology and the significant challenges in translating these innovations from research into clinical practice. Additionally, it emphasizes the limited number of clinical trials utilizing hydrogel therapies for stroke and addresses the associated limitations and constraints, underscoring the need for further research in this field.
Collapse
Affiliation(s)
- Alexandra-Daniela Rotaru-Zăvăleanu
- Department of Epidemiology, University of Medicine and Pharmacy of Craiova, 2-4 Petru Rares Str., 200349 Craiova, Romania;
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Venera Cristina Dinescu
- Department of Health Promotion and Occupational Medicine, University of Medicine and Pharmacy of Craiova, 2–4 Petru Rares Str., 200349 Craiova, Romania
| | - Madalina Aldea
- Psychiatry Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Andrei Gresita
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 115680, USA
| |
Collapse
|
5
|
Hu J, Hu Y, Kang M, Liu X, Wu B, Wang L, Wei Y, Huang D. Sodium alginate/carboxycellulose/polydopamine composite microspheres for rapid hemostasis of deep irregular wounds. Colloids Surf B Biointerfaces 2024; 238:113905. [PMID: 38593680 DOI: 10.1016/j.colsurfb.2024.113905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/11/2024]
Abstract
Hemostasis of deep irregular wounds is a severe problem in clinical practice. The development of rapid-acting hemostatic agents for deep and irregular wound is urgently needed. Here, sodium alginate/carboxycellulose/polydopamine (SA/CNF/PDA) microspheres was prepared by reverse emulsification and crosslinking with Ca2+, and SA/CNF/PDA composite hemostatic microspheres with porous structure were obtained by freeze-drying. SA/CNF/PDA composite hemostatic microspheres exhibited excellent porosity and water absorption which could rapidly absorb blood on the wound surface. Moreover, SA/CNF/PDA composite microspheres demonstrated remarkable hemostatic capabilities both in vitro and in vivo. It exhibited strong hemostatic performance in models of mouse tail-break and liver damage. Especially in liver injury model, it was completely hemostatic in 95 s, and blood loss (19.3 mg). The hemostatic efficacy of the SA/CNF/PDA composite microspheres was amplified through the stimulation of both exogenous and endogenous coagulation pathways. Therefore, SA/CNF/PDA composite hemostatic microspheres are suitable for rapid hemostasis of deep irregular wounds which are potential rapid hemostatic material for surgical application.
Collapse
Affiliation(s)
- Junjie Hu
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Yinchun Hu
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, PR China.
| | - Min Kang
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Xuanyu Liu
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Baogang Wu
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Lining Wang
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Yan Wei
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, PR China
| | - Di Huang
- Research Center for Nano-Biomaterials & Regenerative Medicine, Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, PR China
| |
Collapse
|
6
|
Bhattacharjee A, Savargaonkar AV, Tahir M, Sionkowska A, Popat KC. Surface modification strategies for improved hemocompatibility of polymeric materials: a comprehensive review. RSC Adv 2024; 14:7440-7458. [PMID: 38433935 PMCID: PMC10906639 DOI: 10.1039/d3ra08738g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
Polymeric biomaterials are a widely used class of materials due to their versatile properties. However, as with all other types of materials used for biomaterials, polymers also have to interact with blood. When blood comes into contact with any foreign body, it initiates a cascade which leads to platelet activation and blood coagulation. The implant surface also has to encounter a thromboinflammatory response which makes the implant integrity vulnerable, this leads to blood coagulation on the implant and obstructs it from performing its function. Hence, the surface plays a pivotal role in the design and application of biomaterials. In particular, the surface properties of biomaterials are responsible for biocompatibility with biological systems and hemocompatibility. This review provides a report on recent advances in the field of surface modification approaches for improved hemocompatibility. We focus on the surface properties of polysaccharides, proteins, and synthetic polymers. The blood coagulation cascade has been discussed and blood - material surface interactions have also been explained. The interactions of blood proteins and cells with polymeric material surfaces have been discussed. Moreover, the benefits as well as drawbacks of blood coagulation on the implant surface for wound healing purposes have also been studied. Surface modifications implemented by other researchers to enhance as well as prevent blood coagulation have also been analyzed.
Collapse
Affiliation(s)
- Abhishek Bhattacharjee
- School of Advanced Material Discovery, Colorado State University Fort Collins CO 80523 USA
| | | | - Muhammad Tahir
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University Gagarina 7 87-100 Torun Poland
| | - Alina Sionkowska
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University Gagarina 7 87-100 Torun Poland
| | - Ketul C Popat
- School of Advanced Material Discovery, Colorado State University Fort Collins CO 80523 USA
- Department of Mechanical Engineering, Colorado State University Fort Collins CO 80523 USA
- Department of Bioengineering, George Mason University Fairfax VA 22030 USA
| |
Collapse
|
7
|
Ren Z, Wang Y, Wu H, Cong H, Yu B, Shen Y. Preparation and application of hemostatic microspheres containing biological macromolecules and others. Int J Biol Macromol 2024; 257:128299. [PMID: 38008144 DOI: 10.1016/j.ijbiomac.2023.128299] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/18/2023] [Accepted: 11/18/2023] [Indexed: 11/28/2023]
Abstract
Bleeding from uncontrollable wounds can be fatal, and the body's clotting mechanisms are unable to control bleeding in a timely and effective manner in emergencies such as battlefields and traffic accidents. For irregular and inaccessible wounds, hemostatic materials are needed to intervene to stop bleeding. Hemostatic microspheres are promising for hemostasis, as their unique structural features can promote coagulation. There is a wide choice of materials for the preparation of microspheres, and the modification of natural macromolecular materials such as chitosan to enhance the hemostatic properties and make up for the deficiencies of synthetic macromolecular materials makes the hemostatic microspheres multifunctional and expands the application fields of hemostatic microspheres. Here, we focus on the hemostatic mechanism of different materials and the preparation methods of microspheres, and introduce the modification methods, related properties and applications (in cancer therapy) for the structural characteristics of hemostatic microspheres. Finally, we discuss the future trends of hemostatic microspheres and research opportunities for developing the next generation of hemostatic microsphere materials.
Collapse
Affiliation(s)
- Zekai Ren
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Yumei Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Han Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
8
|
Betancur MI, Case A, Ilich E, Mehta N, Meehan S, Pogrebivsky S, Keir ST, Stevenson K, Brahma B, Gregory S, Chen W, Ashley DM, Bellamkonda R, Mokarram N. A neural tract-inspired conduit for facile, on-demand biopsy of glioblastoma. Neurooncol Adv 2024; 6:vdae064. [PMID: 38813113 PMCID: PMC11135361 DOI: 10.1093/noajnl/vdae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024] Open
Abstract
Background A major hurdle to effectively treating glioblastoma (GBM) patients is the lack of longitudinal information about tumor progression, evolution, and treatment response. Methods In this study, we report the use of a neural tract-inspired conduit containing aligned polymeric nanofibers (i.e., an aligned nanofiber device) to enable on-demand access to GBM tumors in 2 rodent models. Depending on the experiment, a humanized U87MG xenograft and/or F98-GFP+ syngeneic rat tumor model was chosen to test the safety and functionality of the device in providing continuous sampling access to the tumor and its microenvironment. Results The aligned nanofiber device was safe and provided a high quantity of quality genomic materials suitable for omics analyses and yielded a sufficient number of live cells for in vitro expansion and screening. Transcriptomic and genomic analyses demonstrated continuity between material extracted from the device and that of the primary, intracortical tumor (in the in vivo model). Conclusions The results establish the potential of this neural tract-inspired, aligned nanofiber device as an on-demand, safe, and minimally invasive access point, thus enabling rapid, high-throughput, longitudinal assessment of tumor and its microenvironment, ultimately leading to more informed clinical treatment strategies.
Collapse
Affiliation(s)
| | - Ayden Case
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Ekaterina Ilich
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Nalini Mehta
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Sean Meehan
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Sabrina Pogrebivsky
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Stephen T Keir
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Kevin Stevenson
- Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Barun Brahma
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | - Simon Gregory
- Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Wei Chen
- Center for Genomic and Computational Biology, Duke University, Durham, Georgia, USA
| | - David M Ashley
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA
| | - Ravi Bellamkonda
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Nassir Mokarram
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| |
Collapse
|
9
|
Kung Y, Wu CH, Lin MT, Liao WH, Chen WS, Hsiao MY. Blood-cerebrospinal fluid barrier opening by modified single pulse transcranial focused shockwave. Drug Deliv 2023; 30:97-107. [PMID: 36533878 PMCID: PMC9769131 DOI: 10.1080/10717544.2022.2157068] [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] [Indexed: 12/23/2022] Open
Abstract
Transcranial focused shockwave (FSW) is a novel noninvasive brain stimulation that can open blood-brain barriers (BBB) and blood-cerebrospinal fluid barriers (BCSFB) with a single low-energy (energy flux density 0.03 mJ/mm2) pulse and low-dose microbubbles (2 × 106/kg). Similar to focused ultrasound, FSW deliver highly precise stimulation of discrete brain regions with adjustable focal lengths that essentially covers the whole brain. By opening the BCSFB, it allows for rapid widespread drug delivery to the whole brain by cerebrospinal fluid (CSF) circulation. Although no definite adverse effect or permeant injury was noted in our previous study, microscopic hemorrhage was infrequently observed. Safety concerns remain the major obstacle to further application of FSW in brain. To enhance its applicability, a modified single pulse FSW technique was established that present 100% opening rate but much less risk of adverse effect than previous methods. By moving the targeting area 2.5 mm more superficially on the left lateral ventricle as compared with the previous methods, the microscopic hemorrhage rate was reduced to zero. We systemically examine the safety profiles of the modified FSW-BCSFB opening regarding abnormal behavior and brain injury or hemorrhage 72 hr after 0, 1, and 10 pulses of FSW-treatment. Animal behavior, physiological monitor, and brain MRI were examined and recorded. Brain section histology was examined for hemorrhage, apoptosis, inflammation, oxidative stress related immunohistochemistry and biomarkers. The single pulse FSW group demonstrated no mortality or gross/microscopic hemorrhage (N = 30), and no observable changes in all examined outcomes, while 10 pulses of FSW was found to be associated with microscopic and temporary RBC extravasation (N = 6/30), and abnormal immunohistochemistry biomarkers which showed a trend of recovery at 72 hrs. The results suggest that single pulse low-energy FSW-BCSFB opening is effective, safe and poses minimal risk of injury to brain tissue (Sprague Dawley, SD rats).
Collapse
Affiliation(s)
- Yi Kung
- Department of Biomechatronic Engineering, National Chiayi University, Chiayi City, Taiwan
| | - Chueh-Hung Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Meng-Ting Lin
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Wei-Hao Liao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ming-Yen Hsiao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan,CONTACT Ming-Yen Hsiao
| |
Collapse
|
10
|
Villanueva-Flores F, Garcia-Atutxa I, Santos A, Armendariz-Borunda J. Toward a New Generation of Bio-Scaffolds for Neural Tissue Engineering: Challenges and Perspectives. Pharmaceutics 2023; 15:1750. [PMID: 37376198 DOI: 10.3390/pharmaceutics15061750] [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: 05/09/2023] [Revised: 06/04/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Neural tissue engineering presents a compelling technological breakthrough in restoring brain function, holding immense promise. However, the quest to develop implantable scaffolds for neural culture that fulfill all necessary criteria poses a remarkable challenge for material science. These materials must possess a host of desirable characteristics, including support for cellular survival, proliferation, and neuronal migration and the minimization of inflammatory responses. Moreover, they should facilitate electrochemical cell communication, display mechanical properties akin to the brain, emulate the intricate architecture of the extracellular matrix, and ideally allow the controlled release of substances. This comprehensive review delves into the primary requisites, limitations, and prospective avenues for scaffold design in brain tissue engineering. By offering a panoramic overview, our work aims to serve as an essential resource, guiding the creation of materials endowed with bio-mimetic properties, ultimately revolutionizing the treatment of neurological disorders by developing brain-implantable scaffolds.
Collapse
Affiliation(s)
- Francisca Villanueva-Flores
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Chihuahua, Av. Heroico Colegio Militar 4700, Nombre de Dios, Chihuahua 31300, Chihuahua, Mexico
| | - Igor Garcia-Atutxa
- Máster en Bioinformática y Bioestadística, Universitat Oberta de Catalunya, Rambla del Poblenou, 156, 08018 Barcelona, Spain
| | - Arturo Santos
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Guadalajara, Av. Gral Ramón Corona No 2514, Colonia Nuevo México, Zapopan 45201, Jalisco, Mexico
| | - Juan Armendariz-Borunda
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Guadalajara, Av. Gral Ramón Corona No 2514, Colonia Nuevo México, Zapopan 45201, Jalisco, Mexico
- Instituto de Biología Molecular en Medicina y Terapia Génica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
11
|
de Liyis BG, Sutedja JC, Kesuma PMI, Liyis S, Widyadharma IPE. A review of literature on Compound 21-loaded gelatin nanoparticle: a promising nose-to-brain therapy for multi-infarct dementia. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2023. [DOI: 10.1186/s41983-023-00621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
AbstractMulti-infarct dementia (MID) is described as a chronic progressive decline in cortical cognitive function due to the occurrence of multiple infarcts in the cerebral vascularization throughout the gray and white matter. Current therapies of MID mostly focus only on slowing down MID progression and symptomatic medications. A novel therapy which is able to provide both preventive and curative properties for MID is of high interest. The purpose of this review is to identify the potential of Compound 21 (C21) gelatin nanoparticle through the nose-to-brain route as therapy for MID. C21, an angiotensin II type 2 receptor (AT2R) agonist, has shown to reduce the size of cerebral infarct in rodent models, resulting in the preservation and improvement of overall cognitive function and prevention of secondary neurodegenerative effects. It is also shown that C21 decreases neuronal apoptosis, improves damaged axons, and encourage synapse development. The challenge remains in preventing systemic AT2R activation and increasing its low oral bioavailability which can be overcome through nose-to-brain administration of C21. Nose-to-brain drug delivery of C21 significantly increases drug efficiency and limits C21 exposure in order to specifically target the multiple infarcts located in the cerebral cortex. Adhering C21 onto gelatin nanoparticles may enable longer contact time with the olfactory and the trigeminal nerve endings, increasing the potency of C21. In summary, treatment of C21 gelatin nanoparticle through nose-to-brain delivery shows high potential as therapy for vascular dementia. However, clinical trials must be further studied in order to test the safety and efficacy of C21.
Collapse
|
12
|
Mohammed FS, Omay SB, Sheth KN, Zhou J. Nanoparticle-based drug delivery for the treatment of traumatic brain injury. Expert Opin Drug Deliv 2023; 20:55-73. [PMID: 36420918 PMCID: PMC9983310 DOI: 10.1080/17425247.2023.2152001] [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: 08/09/2022] [Revised: 10/10/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Traumatic brain injuries (TBIs) impact the breadth of society and remain without any approved pharmacological treatments. Despite successful Phase II clinical trials, the failure of many Phase III clinical trials may be explained by insufficient drug targeting and retention, preventing the proper attainment of an observable dosage threshold. To address this challenge, nanoparticles can be functionalized to protect pharmacological payloads, improve targeted drug delivery to sites of injury, and can be combined with supportive scaffolding to improve secondary outcomes. AREAS COVERED This review briefly covers the pathophysiology of TBIs and their subtypes, the current pre-clinical and clinical management strategies, explores the common models of focal, diffuse, and mixed traumatic brain injury employed in experimental animals, and surveys the existing literature on nanoparticles developed to treat TBIs. EXPERT OPINION Nanoparticles are well suited to improve secondary outcomes as their multifunctionality and customizability enhance their potential for efficient targeted delivery, payload protection, increased brain penetration, low off-target toxicity, and biocompatibility in both acute and chronic timescales.
Collapse
Affiliation(s)
- Farrah S. Mohammed
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| | - Sacit Bulent Omay
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
| | - Kevin N. Sheth
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
- Department of Neurology, Yale University, New Haven, Connecticut, USA
| | - Jiangbing Zhou
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Department of Neurosurgery, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Wu SD, Dai NT, Liao CY, Kang LY, Tseng YW, Hsu SH. Planar-/Curvilinear-Bioprinted Tri-Cell-Laden Hydrogel for Healing Irregular Chronic Wounds. Adv Healthc Mater 2022; 11:e2201021. [PMID: 35758924 DOI: 10.1002/adhm.202201021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Indexed: 01/24/2023]
Abstract
Chronic cutaneous wounds from tissue trauma or extensive burns can impair skin barrier function and cause severe infection. Fabrication of a customizable tissue-engineered skin is a promising strategy for regeneration of uneven wounds. Herein, a planar-/curvilinear-bioprintable hydrogel is developed to produce tissue-engineered skin and evaluated in rat models of chronic and irregular wounds. The hydrogel is composed of biodegradable polyurethane (PU) and gelatin. The hydrogel laden with cells displays good 3D printability and structure stability. The circular wounds of normal and diabetes mellitus (DM) rats treated with planar-printed tri-cell-laden (fibroblasts, keratinocytes, and endothelial progenitor cells (EPCs)) hydrogel demonstrate full reepithelization and dermal repair as well as large amounts of neovascularization and collagen production after 28 days. Furthermore, the curvilinear module is fabricated based on the corresponding wound topography for curvilinear-bioprinting of the irregular tissue-engineered skin. The large and irregular rat skin wounds treated with curvilinear-printed tri-cell-laden hydrogel demonstrate full repair after 28 days. This planar-/curvilinear-bioprintable tri-cell-laden hydrogel shows great potential for customized biofabrication in skin tissue engineering.
Collapse
Affiliation(s)
- Shin-Da Wu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan
| | - Niann-Tzyy Dai
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Yaug Liao
- Department of Mechanical Engineering, National Central University, Taoyuan, 32001, Taiwan
| | - Lan-Ya Kang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Wen Tseng
- Department of Mechanical Engineering, National Central University, Taoyuan, 32001, Taiwan
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan.,Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
14
|
Wang M, Hu J, Ou Y, He X, Wang Y, Zou C, Jiang Y, Luo F, Lu D, Li Z, Li J, Tan H. Shape-Recoverable Hyaluronic Acid-Waterborne Polyurethane Hybrid Cryogel Accelerates Hemostasis and Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2022; 14:17093-17108. [PMID: 35380771 DOI: 10.1021/acsami.2c01310] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Wound dressings that promote quick hemostasis and are highly efficient in healing wounds are urgently needed to meet the increase in clinical demands worldwide. Herein, a dihydrazide-modified waterborne biodegradable polyurethane emulsion (PU-ADH) and oxidized hyaluronic acid (OHA) were autonomously cross-linked to form a hybrid hyaluronic acid-polyurethane (HA-PU) cryogel by hydrazone bonding at -20 °C. Through its specific macroporous structure (which is approximately 220 μm) constructed by aggregated PU-ADH particles and long-chain OHA, a dried cryogel can have a dramatically compressed volume (1/7 of its original volume) with stable fixation, and it can swell rapidly by absorbing water or blood to approximately 22 and 16 times its dried weight, respectively, in a few minutes. This instantaneous shape-recovering ability favors fast hemostasis in minimally invasive surgery. Moreover, this cryogel is superior to gauze, has excellent biocompatibility, and quickly coagulates blood (in approximately 2 min) by activating the endogenous coagulation system. Comparably, an injectable HA-PU hydrogel with the same components as the HA-PU cryogel was prepared at room temperature, and it exhibited good self-healing properties. An in vivo evaluation of a rat liver hemostasis model and rat skin defect model revealed that the cryogel in fast hemostasis has great potential and superior wound-healing abilities, decreases immune inflammation, and promotes the regeneration of angiogenesis and hair follicles. Consequently, this work proposes a versatile method for constructing biodegradable hybrid cryogels via autonomous cross-linking between synthesized polymer emulsions and natural polymers. The hybrid cryogels demonstrated great potential for applications as high-performance wound dressings.
Collapse
Affiliation(s)
- Min Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Juanjuan Hu
- Department of Otorhinolaryngology, Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yangcen Ou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Xueling He
- Laboratory Animal Center of Sichuan University, Chengdu 610207, China
| | - Yanjun Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Chenyu Zou
- Department of Otorhinolaryngology, Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanlin Jiang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Dan Lu
- Department of Otorhinolaryngology, Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Jiehua Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
15
|
Creative transformation of biomedical polyurethanes: from biostable tubing to biodegradable smart materials. JOURNAL OF POLYMER RESEARCH 2022. [DOI: 10.1007/s10965-022-02919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
16
|
Ghimire S, Sarkar P, Rigby K, Maan A, Mukherjee S, Crawford KE, Mukhopadhyay K. Polymeric Materials for Hemostatic Wound Healing. Pharmaceutics 2021; 13:2127. [PMID: 34959408 PMCID: PMC8708336 DOI: 10.3390/pharmaceutics13122127] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 02/04/2023] Open
Abstract
Hemorrhage is one of the greatest threats to life on the battlefield, accounting for 50% of total deaths. Nearly 86% of combat deaths occur within the first 30 min after wounding. While external wound injuries can be treated mostly using visual inspection, abdominal or internal hemorrhages are more challenging to treat with regular hemostatic dressings because of deep wounds and points of injury that cannot be located properly. The need to treat trauma wounds from limbs, abdomen, liver, stomach, colon, spleen, arterial, venous, and/or parenchymal hemorrhage accompanied by severe bleeding requires an immediate solution that the first responders can apply to reduce rapid exsanguinations from external wounds, including in military operations. This necessitates the development of a unique, easy-to-use, FDA-approved hemostatic treatment that can deliver the agent in less than 30 s and stop bleeding within the first 1 to 2 min at the point of injury without application of manual pressure on the wounded area.
Collapse
Affiliation(s)
- Suvash Ghimire
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA; (S.G.); (P.S.); (K.R.); (A.M.); (S.M.)
| | - Pritha Sarkar
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA; (S.G.); (P.S.); (K.R.); (A.M.); (S.M.)
| | - Kasey Rigby
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA; (S.G.); (P.S.); (K.R.); (A.M.); (S.M.)
| | - Aditya Maan
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA; (S.G.); (P.S.); (K.R.); (A.M.); (S.M.)
- Department of Chemistry, University of Central Florida, Orlando, FL 32816, USA
| | - Santanu Mukherjee
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA; (S.G.); (P.S.); (K.R.); (A.M.); (S.M.)
| | - Kaitlyn E. Crawford
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA; (S.G.); (P.S.); (K.R.); (A.M.); (S.M.)
- Department of Chemistry, University of Central Florida, Orlando, FL 32816, USA
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816, USA
- Biionix Cluster, University of Central Florida, Orlando, FL 32816, USA
| | - Kausik Mukhopadhyay
- Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA; (S.G.); (P.S.); (K.R.); (A.M.); (S.M.)
| |
Collapse
|
17
|
Roy A, Guha Ray P, Manna K, Banerjee C, Dhara S, Pal S. Poly( N-vinyl imidazole) Cross-Linked β-Cyclodextrin Hydrogel for Rapid Hemostasis in Severe Renal Arterial Hemorrhagic Model. Biomacromolecules 2021; 22:5256-5269. [PMID: 34755513 DOI: 10.1021/acs.biomac.1c01174] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A unique facile process has been adopted for fast assembly of a poly(N-vinyl imidazole) cross-linked β-cyclodextrin hydrogel through microwave-assisted free radical polymerization, using N,N'-methylenebis(acrylamide) cross-linker. The copolymer possesses positive surface charge, one of the characteristic properties of an ideal hemostatic hydrogel. The functionalized imidazole-based hydrogel demonstrates rapid, superior blood coagulation kinetics under in vitro and in vivo conditions. On application to a major renal arterial hemorrhagic model, this hydrogel shows better blood clotting kinetics, leading to complete hemostasis in as few as ∼144 ± 7 s. Additionally, 350 μL of whole blood was clotted instantly, in ∼35 s, and therefore, reinforcing its hemostatic potential. The hydrogel demonstrates excellent biocompatibility, when seeded with human dermal fibroblast cells, retaining the native property of its predecessor. In addition, the hydrogel presents excellent hemocompatibility when tested with whole blood with the highest hemolytic ratio of 1.07 ± 0.05%. Moreover, it also demonstrates potential as a carrier for sustained release of an anesthetic drug, lidocaine hydrochloride monohydrate (∼83% in 24 h). The rapid hemostatic behavior of the hydrogel is coupled with its cytocompatibility and hemocompatibilty properties along with controlled drug release characteristics. These behaviors evidently demonstrate it to be an excellent alternative for a superior hemostatic material for severe hemorrhagic conditions.
Collapse
Affiliation(s)
- Arpita Roy
- Department of Chemistry, Indian Institute of Technology (ISM), Dhanbad 826004, India
| | - Preetam Guha Ray
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, India
| | - Kalipada Manna
- Department of Chemistry, Indian Institute of Technology (ISM), Dhanbad 826004, India
| | - Chiranjib Banerjee
- Department of Environmental Science & Engineering, Indian Institute of Technology (ISM), Dhanbad 826004, India
| | - Santanu Dhara
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, India
| | - Sagar Pal
- Department of Chemistry, Indian Institute of Technology (ISM), Dhanbad 826004, India
| |
Collapse
|