1
|
Ale T, Ale T, Baker KJ, Zuniga KM, Hutcheson J, Lavik E. Delivery of Tempol from Polyurethane Nanocapsules to Address Oxidative Stress Post-Injury. Bioconjug Chem 2025; 36:146-151. [PMID: 39921627 DOI: 10.1021/acs.bioconjchem.4c00360] [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: 02/10/2025]
Abstract
Traumatic brain injuries (TBIs) result in significant morbidity and mortality due to the cascade of secondary injuries involving oxidative stress and neuroinflammation. The development of effective therapeutic strategies to mitigate these effects is critical. This study explores the fabrication and characterization of polyurethane nanocapsules for the sustained delivery of Tempol, a potent antioxidant. The nanocapsules were designed to extend the release of Tempol over a 30-day period, addressing the prolonged oxidative stress observed post-TBI. Tempol-loaded polyurethane nanocapsules were synthesized using interfacial polymerization and nanoemulsion techniques. Two generations of nanocapsules were produced, differing in Tempol loading and PEGylation levels. The first generation, with lower Tempol loading, exhibited an average size of 159.8 ± 12.61 nm and a Z-average diameter of 771.9 ± 87.95 nm. The second generation, with higher Tempol loading, showed an average size of 141.4 ± 6.13 nm and a Z-average diameter of 560.7 ± 171.1 nm. The zeta potentials were -18.9 ± 5.02 mV and -11.9 ± 3.54 mV for the first and second generations, respectively. Both generations demonstrated the presence of urethane linkages, confirmed by Fourier Transform Infrared Spectroscopy (FTIR). Loading studies revealed Tempol concentrations of 61.94 ± 3.04 μg/mg for the first generation and 77.61 ± 3.04 μg/mg for the second generation nanocapsules. Release profiles indicated an initial burst followed by a sustained, nearly linear release over 30 days. The higher PEGylation in the second generation nanocapsules is advantageous for intravenous administration, potentially enhancing their therapeutic efficacy in TBI treatment. This study demonstrates the feasibility of using polyurethane nanocapsules for the prolonged delivery of Tempol, offering a promising approach to manage oxidative stress and improve outcomes in TBI patients. Future work will include testing these nanocapsules in vivo to determine their potential at modulating recovery from TBI.
Collapse
Affiliation(s)
- Temitope Ale
- University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Tolulope Ale
- University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Kimberly J Baker
- Defense Health Agency, United States Air Force 59th Medical Wing, Surgical and Technological Advancements for Traumatic Injury in Combat (STATIC), JBSA-Lackland, Texas 78236, United States
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, Texas 78249, United States
| | - Kameel M Zuniga
- Defense Health Agency, United States Air Force 59th Medical Wing, Surgical and Technological Advancements for Traumatic Injury in Combat (STATIC), JBSA-Lackland, Texas 78236, United States
| | - Jack Hutcheson
- Defense Health Agency, United States Air Force 59th Medical Wing, Surgical and Technological Advancements for Traumatic Injury in Combat (STATIC), JBSA-Lackland, Texas 78236, United States
| | - Erin Lavik
- University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland 20850, United States
| |
Collapse
|
2
|
Farrokhi T, Gkikas M. NanoGraphene Clot: A New Fibrinogen-Mimic Hemostatic Material. ACS APPLIED MATERIALS & INTERFACES 2024; 16:34783-34797. [PMID: 38949260 DOI: 10.1021/acsami.4c09828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Trauma is the leading cause of death for adults under the age of 44. Internal bleeding remains a significant challenge in medical emergencies, necessitating the development of effective hemostatic materials that could be administered by paramedics before a patient is in the hospital and treated by surgeons. In this study, we introduce a graphene oxide (GO)-based PEGylated synthetic hemostatic nanomaterial with an average size of 211 ± 83 nm designed to target internal bleeding by mimicking the role of fibrinogen. Functionalization of GO-g-PEG with peptides derived from the α-chain of fibrinogen, such as GRGDS, or the γ-chain of fibrinogen, such as HHLGGAKQAGDV:H12, was achieved with peptide loadings of 72 ± 6 and 68 ± 15 μM, respectively. In vitro studies with platelet-rich plasma (PRP) under confinement demonstrated aggregation enhancement of 39 and 24% for GO-g-PEG-GRGDS and GO-g-PEG-H12, respectively, compared to buffer, while adenosine diphosphate (ADP) alone induced a 5% aggregation. Compared to the same materials in the absence of ADP, GO-g-PEG-GRGDS achieved a 47% aggregation enhancement, while GO-g-PEG-H12 a 25% enhancement. This is particularly important for injectable hemostats and highlights the fact that our nanographene-based materials can only act as hemostats in the presence of agonists, reducing the possibility of unwanted clotting during circulation. Further studies on collagen-coated wells under dynamic flow revealed statistically significant augmentation of PRP fluorescence signal using GRGDS- or H12-coated GO-g-PEG compared to controls. Hemolysis studies showed <1% lysis of red blood cells (RBCs) at the highest PEGylated nanographene concentration. Finally, whole human blood coagulation studies reveal faster and more pronounced clotting using our nanohemostats vs PBS control from 3 min and below (blood is clotted with 10% CaCl2 within 4-5 min), with the biggest differences to be shown at 2 and 1 min. At 1 min, the clot weight was found to be ∼45% of that between 4 and 5 min, while no clot was formed in PBS-treated blood. Reduction of CaCl2 to 5 and 3%, or utilization of prostaglandin E1, an anticoagulant, still leads to clots but of smaller weight. The findings highlight the potential of our fibrinogen-mimic PEGylated nanographene as a promising non-hemolytic injectable scaffold for targeting internal bleeding, offering insights into its platelet aggregation capabilities under confinement and under dynamic flow as well as its pronounced coagulation abilities.
Collapse
Affiliation(s)
- Tannaz Farrokhi
- Department of Chemistry, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| | - Manos Gkikas
- Department of Chemistry, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| |
Collapse
|
3
|
Srinivasan AJ, Secunda ZA, Mota-Alvidrez RI, Luc NF, Disharoon D, Traylor B, Pawlowski CL, Brown JB, Bruckman MA, Sen Gupta A, Neal MD. Platelet-inspired synthetic nanoparticles improve hemostasis and hemodynamics in a rabbit model of abdominal hemorrhage. J Trauma Acute Care Surg 2024; 96:101-108. [PMID: 38057963 PMCID: PMC10746291 DOI: 10.1097/ta.0000000000003938] [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] [Indexed: 12/08/2023]
Abstract
BACKGROUND Early platelet transfusion is associated with reduced mortality in traumatic hemorrhage. However, platelet usage is severely limited because of the challenges of donor availability, platelet portability, and storage. Here, we report on a bioinspired synthetic platelet (SP) nanoconstruct that utilizes liposome surface-decoration with peptides that mimic injury site-specific platelet adhesion to von Willebrand Factor and collagen, and fibrinogen-mediated platelet aggregation. Synthetic platelet has previously shown promising hemostatic outcomes in vitro and in vivo. Here, we evaluated hemostasis and hemodynamic effects of SP in a rabbit model of abdominal hemorrhage. METHODS Twenty-three adult male New Zealand white rabbits (2.5-3.5 kg) were treated with either buffer, control particles (CPs), or SP. Under general anesthesia with invasive monitoring, rabbits underwent laparotomy with combined splenic and hepatic injury. Hemodynamics were monitored for 30 minutes and blood loss was quantified. Blood counts, aggregometry, catecholamine and platelet factor 4 (PF4) assays were performed at multiple timepoints. Analysis used analysis of variance and post hoc Tukey testing with α = 0.05. RESULTS Rabbits in the SP (n = 7) group had significantly lower weight-normalized blood loss compared with both buffer (n = 8) and CP (n = 8) animals (21.1 vs. 33.2 vs. 40.4 g/kg, p < 0.001). Synthetic platelet-treated animals had higher systolic blood pressure area under curve compared with buffer- and CP-treated animals (1567 vs. 1281 vs. 1109 mm Hg*min, p = 0.006), although post hoc differences were only significant for the SP/CP comparison ( p = 0.005). Platelet counts, catecholamine levels, PF4, and aggregometry were similar between groups. CONCLUSION Synthetic platelet treatment significantly reduced blood loss and improved hemodynamics in a rabbit abdominal hemorrhage model. Synthetic platelet has potential as an intravenous hemostatic platelet surrogate with donor-independent availability and scalable manufacture.
Collapse
Affiliation(s)
- Amudan J. Srinivasan
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh PA 15213
| | - Zachary A. Secunda
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh PA 15213
| | - Roberto I. Mota-Alvidrez
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh PA 15213
| | - Norman F. Luc
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Dante Disharoon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | | | | | - Joshua B. Brown
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh PA 15213
| | | | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Matthew D. Neal
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh PA 15213
| |
Collapse
|
4
|
Hong C, He Y, Bowen PA, Belcher AM, Olsen BD, Hammond PT. Engineering a Two-Component Hemostat for the Treatment of Internal Bleeding through Wound-Targeted Crosslinking. Adv Healthc Mater 2023; 12:e2202756. [PMID: 37017403 PMCID: PMC10964210 DOI: 10.1002/adhm.202202756] [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: 10/25/2022] [Revised: 03/01/2023] [Indexed: 04/06/2023]
Abstract
Primary hemostasis (platelet plug formation) and secondary hemostasis (fibrin clot formation) are intertwined processes that occur upon vascular injury. Researchers have sought to target wounds by leveraging cues specific to these processes, such as using peptides that bind activated platelets or fibrin. While these materials have shown success in various injury models, they are commonly designed for the purpose of treating solely primary or secondary hemostasis. In this work, a two-component system consisting of a targeting component (azide/GRGDS PEG-PLGA nanoparticles) and a crosslinking component (multifunctional DBCO) is developed to treat internal bleeding. The system leverages increased injury accumulation to achieve crosslinking above a critical concentration, addressing both primary and secondary hemostasis by amplifying platelet recruitment and mitigating plasminolysis for greater clot stability. Nanoparticle aggregation is measured to validate concentration-dependent crosslinking, while a 1:3 azide/GRGDS ratio is found to increase platelet recruitment, decrease clot degradation in hemodiluted environments, and decrease complement activation. Finally, this approach significantly increases survival relative to the particle-only control in a liver resection model. In light of prior successes with the particle-only system, these results emphasize the potential of this technology in aiding hemostasis and the importance of a holistic approach in engineering new treatments for hemorrhage.
Collapse
Affiliation(s)
- Celestine Hong
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Institute for Soldier NanotechnologiesMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Yanpu He
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Porter A. Bowen
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Angela M. Belcher
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Bradley D. Olsen
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Institute for Soldier NanotechnologiesMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Paula T. Hammond
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Institute for Soldier NanotechnologiesMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
5
|
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
|
6
|
Li XF, Lu P, Jia HR, Li G, Zhu B, Wang X, Wu FG. Emerging materials for hemostasis. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214823] [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]
|
7
|
Chen Z, Han L, Meng G, Li H, Shan C, Du G, Li M. Intravenous Hemostats: Foundation, Targeting, and Controlled-Release. Bioconjug Chem 2022; 33:2269-2289. [PMID: 36404605 DOI: 10.1021/acs.bioconjchem.2c00492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Uncontrollable blood loss is the greatest cause of mortality in prehospital patients and the main source of disability and death in hospital care. Compared with external hemostats, intravenous hemostats are more appropriate for preventing and treating uncontrolled bleeding in vivo and large bleeding on the body surface. This Review initially establishes intravenous hemostats' response basis, including the coagulation mechanism, fibrinolytic pathway, and protein corona. Second, the study of advancement of intravenous hemostat targeting was expanded from two perspectives, cellular hemostatic agents and synthetic hemostatic agents. Meanwhile, after discussing the progress of controlled-release intravenous hemostats with platelets as the stimuli, this Review offers insight into the possibility of controlled-release intravenous hemostats with microenvironment as the stimuli, combining the studies of controlled-release targeted thrombolysis.
Collapse
Affiliation(s)
- Zihao Chen
- Department of Special Operations Medicine, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Lei Han
- Department of Special Operations Medicine, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Guo Meng
- Department of Special Operations Medicine, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Huaiyong Li
- Department of Special Operations Medicine, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Chao Shan
- Department of Special Operations Medicine, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Ge Du
- Department Of Geriatric Rehabilitation Center, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Beijing 100144, China
| | - Minggao Li
- Department of Special Operations Medicine, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| |
Collapse
|
8
|
Jiang W, Wu Z, Gao Z, Wan M, Zhou M, Mao C, Shen J. Artificial Cells: Past, Present and Future. ACS NANO 2022; 16:15705-15733. [PMID: 36226996 DOI: 10.1021/acsnano.2c06104] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Artificial cells are constructed to imitate natural cells and allow researchers to explore biological process and the origin of life. The construction methods for artificial cells, through both top-down or bottom-up approaches, have achieved great progress over the past decades. Here we present a comprehensive overview on the development of artificial cells and their properties and applications. Artificial cells are derived from lipids, polymers, lipid/polymer hybrids, natural cell membranes, colloidosome, metal-organic frameworks and coacervates. They can be endowed with various functions through the incorporation of proteins and genes on the cell surface or encapsulated inside of the cells. These modulations determine the properties of artificial cells, including producing energy, cell growth, morphology change, division, transmembrane transport, environmental response, motility and chemotaxis. Multiple applications of these artificial cells are discussed here with a focus on therapeutic applications. Artificial cells are used as carriers for materials and information exchange and have been shown to function as targeted delivery systems of personalized drugs. Additionally, artificial cells can function to substitute for cells with impaired function. Enzyme therapy and immunotherapy using artificial cells have been an intense focus of research. Finally, prospects of future development of cell-mimic properties and broader applications are highlighted.
Collapse
Affiliation(s)
- Wentao Jiang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ziyu Wu
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Zheng Gao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Min Zhou
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
9
|
Girish A, Jolly K, Alsaadi N, de la Fuente M, Recchione A, An R, Disharoon D, Secunda Z, Raghunathan S, Luc NF, Desai C, Knauss E, Han X, Hu K, Wang H, Sekhon UDS, Rohner N, Gurkan UA, Nieman M, Neal MD, Sen Gupta A. Platelet-Inspired Intravenous Nanomedicine for Injury-Targeted Direct Delivery of Thrombin to Augment Hemostasis in Coagulopathies. ACS NANO 2022; 16:16292-16313. [PMID: 35916497 PMCID: PMC10195184 DOI: 10.1021/acsnano.2c05306] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Severe hemorrhage associated with trauma, surgery, and congenital or drug-induced coagulopathies can be life-threatening and requires rapid hemostatic management via topical, intracavitary, or intravenous routes. For injuries that are not easily accessible externally, intravenous hemostatic approaches are needed. The clinical gold standard for this is transfusion of blood products, but due to donor dependence, specialized storage requirements, high risk of contamination, and short shelf life, blood product use faces significant challenges. Consequently, recent research efforts are being focused on designing biosynthetic intravenous hemostats, using intravenous nanoparticles and polymer systems. Here we report on the design and evaluation of thrombin-loaded injury-site-targeted lipid nanoparticles (t-TLNPs) that can specifically localize at an injury site via platelet-mimetic anchorage to the von Willebrand factor (vWF) and collagen and directly release thrombin via diffusion and phospholipase-triggered particle destabilization, which can locally augment fibrin generation from fibrinogen for hemostatic action. We evaluated t-TLNPs in vitro in human blood and plasma, where hemostatic defects were created by platelet depletion and anticoagulation. Spectrophotometric studies of fibrin generation, rotational thromboelastometry (ROTEM)-based studies of clot viscoelasticity, and BioFlux-based real-time imaging of fibrin generation under simulated vascular flow conditions confirmed that t-TLNPs can restore fibrin in hemostatic dysfunction settings. Finally, the in vivo feasibility of t-TLNPs was tested by prophylactic administration in a tail-clip model and emergency administration in a liver-laceration model in mice with induced hemostatic defects. Treatment with t-TLNPs was able to significantly reduce bleeding in both models. Our studies demonstrate an intravenous nanomedicine approach for injury-site-targeted direct delivery of thrombin to augment hemostasis.
Collapse
Affiliation(s)
- Aditya Girish
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Ketan Jolly
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Nijmeh Alsaadi
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15123, United States
| | - Maria de la Fuente
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Arielle Recchione
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Ran An
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Dante Disharoon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Zachary Secunda
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15123, United States
| | - Shruti Raghunathan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Norman F Luc
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Cian Desai
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Elizabeth Knauss
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Xu Han
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Keren Hu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Hanyang Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Ujjal Didar Singh Sekhon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Nathan Rohner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Umut A Gurkan
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Marvin Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Matthew D Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15123, United States
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
10
|
Beyer M, France J, Nagaraja TN, Lavik EB, Knight RA, Lewandowski CA, Miller JB. Unaffected ex vivo clotting cascade by experimental hemostatic nanoparticles when introduced in the presence of recombinant tissue plasminogen activator. Brain Circ 2022; 8:228-231. [PMID: 37181845 PMCID: PMC10167850 DOI: 10.4103/bc.bc_45_22] [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: 07/08/2022] [Revised: 09/02/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022] Open
Abstract
CONTEXT Hemostatic nanoparticles (hNPs) have shown efficacy in decreasing intracerebral hemorrhage (ICH) in animal models and are suggested to be of use to counter tissue plasminogen activator (tPA)-induced acute ICH. AIMS The objective of this study was to test the ability of an hNP preparation to alter the clotting properties of blood exposed to tPA ex vivo. MATERIALS AND METHODS Fresh blood samples were obtained from normal male Sprague-Dawley rats (~300 g; n = 6) and prepared for coagulation assays by thromboelastography (TEG) methods. Samples were untreated, exposed to tPA, or exposed to tPA and then to hNP. TEG parameters included reaction time (R, time in minutes elapsed from test initiation to initial fibrin formation), coagulation time (K, time in minutes from R until initial clot formation), angle (α, a measure in degrees of the rate of clot formation), maximum amplitude (MA, the point when the clot reaches its MA in mm), lysis at 30 min after MA (LY30, %), and clot strength (G, dynes/cm2), an index of clot strength. STATISTICAL ANALYSIS USED Kruskal-Wallis test was employed to compare TEG parameters measured for untreated control samples versus those exposed to tPA and to compare tPA-exposed samples to samples treated with tPA + hNPs. Significances were inferred at P ≤ 0.05. RESULTS Compared to untreated samples, tPA-treated samples showed a trend toward decreased angle and G suggesting potentially clot formation rate and clot strength. The addition of hNP did not affect any of these or other measured indices. CONCLUSIONS The data demonstrated no hemostatic effects when the hNP was used in the presence of tPA. The lack of change in any of the TEG parameters measured in the present study may indicate limitations of the hNPs to reverse the thrombolytic cascade initiated by tPA.
Collapse
Affiliation(s)
- Margaret Beyer
- Department of Emergency Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - John France
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, MI, USA
| | | | - Erin B. Lavik
- Department of Chemical, Biochemical, and Environmental Engineering, University of Maryland, Baltimore, MD, USA
| | | | | | - Joseph B. Miller
- Department of Emergency Medicine, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
11
|
Maisha N, Kulkarni C, Pandala N, Zilberberg R, Schaub L, Neidert L, Glaser J, Cannon J, Janeja V, Lavik EB. PEGylated Polyester Nanoparticles Trigger Adverse Events in a Large Animal Model of Trauma and in Naı̈ve Animals: Understanding Cytokine and Cellular Correlations with These Events. ACS NANO 2022; 16:10566-10580. [PMID: 35822898 DOI: 10.1021/acsnano.2c01993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Intravenously infusible nanoparticles to control bleeding have shown promise in rodents, but translation into preclinical models has been challenging as many of these nanoparticle approaches have resulted in infusion responses and adverse outcomes in large animal trauma models. We developed a hemostatic nanoparticle technology that was screened to avoid one component of the infusion response: complement activation. We administered these hemostatic nanoparticles, control nanoparticles, or saline volume controls in a porcine polytrauma model. While the hemostatic nanoparticles promoted clotting as marked by a decrease in prothrombin time and both the hemostatic nanoparticles and controls did not active complement, in a subset of the animals, hard thrombi were found in uninjured tissues in both the hemostatic and control nanoparticle groups. Using data science methods that allow one to work across heterogeneous data sets, we found that the presence of these thrombi correlated with changes in IL-6, INF-alpha, lymphocytes, and neutrophils. While these findings might suggest that this formulation would not be a safe one for translation for trauma, they provide guidance for developing screening tools to make nanoparticle formulations in the complex milieux of trauma as well as for therapeutic interventions more broadly. This is important as we look to translate intravenously administered nanoparticle formulations for therapies, particularly considering the vascular changes seen in a subset of patients following COVID-19. We need to understand adverse events like thrombi more completely and screen for these events early to make nanomaterials as safe and effective as possible.
Collapse
Affiliation(s)
| | | | | | | | - Leasha Schaub
- Naval Medical Research Unit-San Antonio, San Antonio, Texas 78234, United States
| | - Leslie Neidert
- Naval Medical Research Unit-San Antonio, San Antonio, Texas 78234, United States
| | - Jacob Glaser
- Naval Medical Research Unit-San Antonio, San Antonio, Texas 78234, United States
| | - Jeremy Cannon
- Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | | | | |
Collapse
|
12
|
Moldovan H, Antoniac I, Gheorghiță D, Safta MS, Preda S, Broască M, Badilă E, Fronea O, Scafa-Udrişte A, Cacoveanu M, Molnar A, Costache VS, Zaharia O. Biomaterials as Haemostatic Agents in Cardiovascular Surgery: Review of Current Situation and Future Trends. Polymers (Basel) 2022; 14:1189. [PMID: 35335519 PMCID: PMC8955858 DOI: 10.3390/polym14061189] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Intraoperative haemostasis is of paramount importance in the practice of cardiovascular surgery. Over the past 70 years, topical haemostatic methods have advanced significantly and today we deal with various haemostatic agents with different properties and different mechanisms of action. The particularity of coagulation mechanisms after extracorporeal circulation, has encouraged the introduction of new types of topic agents to achieve haemostasis, where conventional methods prove their limits. These products have an important role in cardiac, as well as in vascular, surgery, mainly in major vascular procedures, like aortic dissections and aortic aneurysms. This article presents those agents used for topical application and the mechanism of haemostasis and offers general recommendations for their use in the operating room.
Collapse
Affiliation(s)
- Horațiu Moldovan
- Department of Cardiovascular Surgery, Bucharest Clinical Emergency Hospital, 014461 Bucharest, Romania; (M.S.S.); (S.P.); (M.B.); (A.S.-U.); (M.C.)
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.B.); (O.F.); (O.Z.)
| | - Iulian Antoniac
- Faculty of Materials Science and Engineering, Politehnica University of Bucharest, 060042 Bucharest, Romania;
| | - Daniela Gheorghiță
- Faculty of Materials Science and Engineering, Politehnica University of Bucharest, 060042 Bucharest, Romania;
| | - Maria Sabina Safta
- Department of Cardiovascular Surgery, Bucharest Clinical Emergency Hospital, 014461 Bucharest, Romania; (M.S.S.); (S.P.); (M.B.); (A.S.-U.); (M.C.)
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.B.); (O.F.); (O.Z.)
| | - Silvia Preda
- Department of Cardiovascular Surgery, Bucharest Clinical Emergency Hospital, 014461 Bucharest, Romania; (M.S.S.); (S.P.); (M.B.); (A.S.-U.); (M.C.)
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.B.); (O.F.); (O.Z.)
| | - Marian Broască
- Department of Cardiovascular Surgery, Bucharest Clinical Emergency Hospital, 014461 Bucharest, Romania; (M.S.S.); (S.P.); (M.B.); (A.S.-U.); (M.C.)
| | - Elisabeta Badilă
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.B.); (O.F.); (O.Z.)
| | - Oana Fronea
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.B.); (O.F.); (O.Z.)
| | - Alexandru Scafa-Udrişte
- Department of Cardiovascular Surgery, Bucharest Clinical Emergency Hospital, 014461 Bucharest, Romania; (M.S.S.); (S.P.); (M.B.); (A.S.-U.); (M.C.)
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.B.); (O.F.); (O.Z.)
| | - Mihai Cacoveanu
- Department of Cardiovascular Surgery, Bucharest Clinical Emergency Hospital, 014461 Bucharest, Romania; (M.S.S.); (S.P.); (M.B.); (A.S.-U.); (M.C.)
| | - Adrian Molnar
- Faculty of Medicine, Iuliu Hateganu University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania;
- Heart Institute, 400001 Cluj-Napoca, Romania
| | - Victor Sebastian Costache
- Sf. Constantin Hospital, 500388 Brasov, Romania;
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania
| | - Ondin Zaharia
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.B.); (O.F.); (O.Z.)
- Prof.Dr. Theodor Burghele Clinical Hospital, 050659 Bucharest, Romania
| |
Collapse
|
13
|
Hong C, Alser O, Gebran A, He Y, Joo W, Kokoroskos N, Velmahos G, Olsen BD, Hammond PT. Modulating Nanoparticle Size to Understand Factors Affecting Hemostatic Efficacy and Maximize Survival in a Lethal Inferior Vena Cava Injury Model. ACS NANO 2022; 16:2494-2510. [PMID: 35090344 PMCID: PMC9989960 DOI: 10.1021/acsnano.1c09108] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Intravenous nanoparticle hemostats offer a potentially attractive approach to promote hemostasis, in particular for inaccessible wounds such as noncompressible torso hemorrhage (NCTH). In this work, particle size was tuned over a range of <100-500 nm, and its effect on nanoparticle-platelet interactions was systematically assessed using in vitro and in vivo experiments. Smaller particles bound a larger percentage of platelets per mass of particle delivered, while larger particles resulted in higher particle accumulation on a surface of platelets and collagen. Intermediate particles led to the greatest platelet content in platelet-nanoparticle aggregates, indicating that they may be able to recruit more platelets to the wound. In biodistribution studies, smaller and intermediate nanoparticles exhibited longer circulation lifetimes, while larger nanoparticles resulted in higher pulmonary accumulation. The particles were then challenged in a 2 h lethal inferior vena cava (IVC) puncture model, where intermediate nanoparticles significantly increased both survival and injury-specific targeting relative to saline and unfunctionalized particle controls. An increase in survival in the second hour was likewise observed in the smaller nanoparticles relative to saline controls, though no significant increase in survival was observed in the larger nanoparticle size. In conjunction with prior in vitro and in vivo experiments, these results suggest that platelet content in aggregates and extended nanoparticle circulation lifetimes are instrumental to enhancing hemostasis. Ultimately, this study elucidates the role of particle size in platelet-particle interactions, which can be a useful tool for engineering the performance of particulate hemostats and improving the design of these materials.
Collapse
Affiliation(s)
- Celestine Hong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Osaid Alser
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Department of Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts 02144, USA
| | - Anthony Gebran
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Department of Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts 02144, USA
| | - Yanpu He
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wontae Joo
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Nikolaos Kokoroskos
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Department of Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts 02144, USA
| | - George Velmahos
- Division of Trauma, Emergency Surgery and Surgical Critical Care, Department of Surgery, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts 02144, USA
| | - Bradley D. Olsen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Paula T. Hammond
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
14
|
A review of treatments for non-compressible torso hemorrhage (NCTH) and internal bleeding. Biomaterials 2022; 283:121432. [DOI: 10.1016/j.biomaterials.2022.121432] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/26/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022]
|
15
|
Luc NF, Rohner N, Girish A, Sekhon UDS, Neal MD, Gupta AS. Bioinspired artificial platelets: past, present and future. Platelets 2022; 33:35-47. [PMID: 34455908 PMCID: PMC8795470 DOI: 10.1080/09537104.2021.1967916] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Platelets are anucleate blood cells produced from megakaryocytes predominantly in the bone marrow and released into blood circulation at a healthy count of 150,000-400,00 per μL and circulation lifespan of 7-9 days. Platelets are the first responders at the site of vascular injury and bleeding, and participate in clot formation via injury site-specific primary mechanisms of adhesion, activation and aggregation to form a platelet plug, as well as secondary mechanisms of augmenting coagulation via thrombin amplification and fibrin generation. Platelets also secrete various granule contents that enhance these mechanisms for clot growth and stability. The resultant clot seals the injury site to stanch bleeding, a process termed as hemostasis. Due to this critical role, a reduction in platelet count or dysregulation in platelet function is associated with bleeding risks and hemorrhagic complications. These scenarios are often treated by prophylactic or emergency transfusion of platelets. However, platelet transfusions face significant challenges due to limited donor availability, difficult portability and storage, high bacterial contamination risks, and very short shelf life (~5-7 days). These are currently being addressed by a robust volume of research involving reduced temperature storage and pathogen reduction processes on donor platelets to improve shelf-life and reduce contamination, as well as bioreactor-based approaches to generate donor-independent platelets from stem cells in vitro. In parallel, a complementary research field has emerged that involves the design of artificial platelets utilizing biosynthetic particle constructs that functionally emulate various hemostatic mechanisms of platelets. Here, we provide a comprehensive review of the history and the current state-of-the-art artificial platelet approaches, along with discussing the translational opportunities and challenges.
Collapse
Affiliation(s)
- Norman F. Luc
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA
| | - Nathan Rohner
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA
| | - Aditya Girish
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA
| | | | - Matthew D. Neal
- University of Pittsburgh, Pittsburgh Trauma Research Center, Department of Surgery, Pittsburgh, PA 15123, USA
| | - Anirban Sen Gupta
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH 44106, USA
| |
Collapse
|
16
|
|
17
|
Maisha N, Rubenstein M, Bieberich CJ, Lavik E. Getting to the Core of It All: Nanocapsules to Mitigate Infusion Reactions Can Promote Hemostasis and Be a Platform for Intravenous Therapies. NANO LETTERS 2021; 21:9069-9076. [PMID: 34714087 DOI: 10.1021/acs.nanolett.1c02746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
One of the significant challenges to translation of intravenously administered nanomaterials has been complement-mediated infusion reactions which can be lethal. Slow infusions can reduce infusion reactions, but slow infusions are not always possible in applications like controlling bleeding following trauma. Thus, avoiding complement activation and infusion responses is essential to manage bleeding. We identified nanocapsules based on polyurethane as candidates that did not activate C5a and explored their PEGylation and functionalization with the GRGDS peptide to create a new class of hemostatic nanomaterials. Using the clinically relevant rotational thromboelastography (ROTEM), we determined that nanocapsules promote faster clotting than controls and maintain the maximum clot firmness, which is critical for reducing bleeding. Excitingly, these polyurethane-based nanocapsules did not activate complement or the major pro-inflammatory cytokines. This work provides critical evidence for the role of modulating the core material in developing safer nanomedicines for intravenous applications.
Collapse
Affiliation(s)
- Nuzhat Maisha
- University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| | - Michael Rubenstein
- University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| | - Charles J Bieberich
- University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| | - Erin Lavik
- University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| |
Collapse
|
18
|
Zinger A, Soriano S, Baudo G, De Rosa E, Taraballi F, Villapol S. Biomimetic Nanoparticles as a Theranostic Tool for Traumatic Brain Injury. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2100722. [PMID: 34413716 PMCID: PMC8356641 DOI: 10.1002/adfm.202100722] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/08/2021] [Indexed: 05/04/2023]
Abstract
Traumatic brain injury (TBI) triggers both central and peripheral inflammatory responses. Existing pharmacological drugs are unable to effectively and quickly target the brain inflamed regions, setting up a major roadblock towards effective brain trauma treatments. Nanoparticles (NPs) have been used in multiple diseases as drug delivery tools with remarkable success due to their rapid diffusion and specificity in the target organ. Here, leukocyte-based biomimetic NPs are fabricated as a theranostic tool to directly access inflamed regions in a TBI mouse model. This NP systemic delivery is visualized using advanced in vivo imaging techniques, including intravital microscopy and in vivo imaging system. The results demonstrate selective targeting of NPs to the injured brain and increased NPs accumulation among the peripheral organs 24 h after TBI. Interestingly, increased microglial proliferation, decreased macrophage infiltration, and reduced brain lesion following the NPs treatments compared to sham vehicle-treated mice are also found. In summary, the results suggest that NPs represent a promising future theranostic tool for TBI treatment.
Collapse
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Sirena Soriano
- Center for Neuroregeneration and Department of NeurosurgeryHouston Methodist Research InstituteHoustonTX77030USA
| | - Gherardo Baudo
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Enrica De Rosa
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Francesca Taraballi
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Sonia Villapol
- Center for Neuroregeneration and Department of NeurosurgeryHouston Methodist Research InstituteHoustonTX77030USA
- Weill Cornell Medical CollegeNew YorkNY10065USA
| |
Collapse
|
19
|
Platelet Transfusion-Insights from Current Practice to Future Development. J Clin Med 2021; 10:jcm10091990. [PMID: 34066360 PMCID: PMC8125287 DOI: 10.3390/jcm10091990] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Since the late sixties, therapeutic or prophylactic platelet transfusion has been used to relieve hemorrhagic complications of patients with, e.g., thrombocytopenia, platelet dysfunction, and injuries, and is an essential part of the supportive care in high dose chemotherapy. Current and upcoming advances will significantly affect present standards. We focus on specific issues, including the comparison of buffy-coat (BPC) and apheresis platelet concentrates (APC); plasma additive solutions (PAS); further measures for improvement of platelet storage quality; pathogen inactivation; and cold storage of platelets. The objective of this article is to give insights from current practice to future development on platelet transfusion, focusing on these selected issues, which have a potentially major impact on forthcoming guidelines.
Collapse
|
20
|
Cau MF, Strilchuk AW, Kastrup CJ. Nanomedicines for hemorrhage control. J Thromb Haemost 2021; 19:887-891. [PMID: 33694243 DOI: 10.1111/jth.15211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 11/27/2022]
Affiliation(s)
- Massimo F Cau
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Amy W Strilchuk
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Christian J Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Malik A, Rehman FU, Shah KU, Naz SS, Qaisar S. Hemostatic strategies for uncontrolled bleeding: A comprehensive update. J Biomed Mater Res B Appl Biomater 2021; 109:1465-1477. [PMID: 33511753 DOI: 10.1002/jbm.b.34806] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/31/2020] [Accepted: 01/14/2021] [Indexed: 11/10/2022]
Abstract
Uncontrolled bleeding remains the leading cause of morbidity and mortality across the entire macrocosm. It refers to excessive loss of blood that occurs inside of body, due to unsuccessful platelet plug formation at the injury site. It is not only limited to the battlefield, but remains the second leading cause of death amongst the civilians, as a result of traumatic injury. Startlingly, there are no effective treatments currently available, to cater the issue of internal bleeding, even though early intervention is of utmost significance in minimizing the mortality rates associated with it. The fatal issue of uncontrolled bleeding is ineffectively being dealt with the use of pressure dressings, tourniquet, and surgical procedures. This is not a practical approach in combat arenas or in emergency situations, where the traumatic injury inflicted is deep inside the body, and cannot be addressed externally, by the application of topical dressings. This review focuses on the traditional hemostatic agents that are used to augment the process of hemostasis, such as mineral zeolites, chitosan based products, biologically active agents, anti-fibrinolytics, absorbable agents, and albumin and glutaraldehyde, as well as the micro- and nano-based hemostatic agents such as synthocytes, thromboerythrocytes, thrombosomes, and the synthetic platelets.
Collapse
Affiliation(s)
- Annum Malik
- Nanosciences and Technology Department, National Centre for Physics, Quaid-i-Azam University Campus, Islamabad, Pakistan.,Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fiza Ur Rehman
- Nanosciences and Technology Department, National Centre for Physics, Quaid-i-Azam University Campus, Islamabad, Pakistan.,Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | | | - Syeda Sohaila Naz
- Nanosciences and Technology Department, National Centre for Physics, Quaid-i-Azam University Campus, Islamabad, Pakistan
| | - Sara Qaisar
- Nanosciences and Technology Department, National Centre for Physics, Quaid-i-Azam University Campus, Islamabad, Pakistan
| |
Collapse
|
22
|
Ghosh S, Tripathi A, Gayen P, Sinha Roy R. Peptide-based topical agents and intravenous hemostat for rapid hemostasis. RSC Med Chem 2020; 11:1100-1111. [PMID: 33479616 PMCID: PMC7651999 DOI: 10.1039/d0md00204f] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/14/2020] [Indexed: 01/25/2023] Open
Abstract
Traumatic coagulopathy due to severe external injury and internal hemorrhage is life-threatening to accident victims and soldiers on the battlefield, causing considerable number of deaths worldwide. Patients with inherited bleeding disorders (such as haemophilia, von Willebrand disease, inherited qualitative platelet defects, and afibrinogenemia) also contribute to the vast number of deaths due to abnormal bleeding, and these patients are difficult to handle during surgery. Platelets and different plasma proteins play an essential role in blood coagulation and in the maintenance of the body's hemostatic balance. The improper function or deficiency of these factors cause abnormal bleeding. To address such bleeding disorders, external clotting agents (such as extracellular protein-inspired natural and synthetic peptide-based sealants and peptide-functionalized polymer/liposome-based sealants) have been developed by different groups of researchers. The primary focus of this review is to provide molecular insights into the existing biologically inspired peptide-based sealants, highlighting the advantages and limitations of such reported designed sealants to handle blood clotting, and also provide insights into the design of improved next-generation surgical sealants.
Collapse
Affiliation(s)
- Snehasish Ghosh
- Department of Chemical Sciences , Indian Institute of Science Education and Research Kolkata , Mohanpur - 741246 , India
| | - Archana Tripathi
- Department of Biological Sciences , Indian Institute of Science Education and Research Kolkata , Mohanpur - 741246 , India .
| | - Paramita Gayen
- Department of Biological Sciences , Indian Institute of Science Education and Research Kolkata , Mohanpur - 741246 , India .
| | - Rituparna Sinha Roy
- Department of Biological Sciences , Indian Institute of Science Education and Research Kolkata , Mohanpur - 741246 , India .
- Centre for Advanced Functional Materials , Indian Institute of Science Education and Research Kolkata , Mohanpur - 741246 , India
- Centre for Climate and Environmental Studies , Indian Institute of Science Education and Research Kolkata , Mohanpur - 741246 , India
| |
Collapse
|
23
|
Klein MK, Tsihlis ND, Pritts TA, Kibbe MR. Emerging Therapies for Prehospital Control of Hemorrhage. J Surg Res 2020; 248:182-190. [PMID: 31711614 DOI: 10.1016/j.jss.2019.09.070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/09/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The aim of this review was to describe emerging therapies that could serve as a prehospital intervention to slow or stop noncompressible torso hemorrhage in the civilian and military settings. Hemorrhage accounts for 90% of potentially survivable military deaths and 30%-40% of trauma deaths. There is a great need to develop novel therapies to slow or stop noncompressible torso hemorrhage at the scene of the injury. METHODS A comprehensive literature search was performed using PubMed (1966 to present) for therapies not approved by the Food and Drug Administration for noncompressible torso hemorrhage in the prehospital setting. Therapies were divided into compressive versus intravascular injectable therapies. Ease of administration, skill required to use the therapy, safety profile, stability, shelf-life, mortality benefit, and efficacy were reviewed. RESULTS Multiple potential therapies for noncompressible torso hemorrhage are currently under active investigation. These include (1) tamponade therapies, such as gas insufflation and polyurethane foam injection; (2) freeze-dried blood products and alternatives such as lyophilized platelets; (3) nanoscale injectable therapies such as polyethylene glycol nanospheres, polyethylenimine nanoparticles, SynthoPlate, and tissue factor-targeted nanofibers; and (4) other injectable therapies such as polySTAT and adenosine, lidocaine, and magnesium. Although each of these therapies shows great promise at slowing or stopping hemorrhage in animal models of noncompressible hemorrhage, further research is needed to ensure safety and efficacy in humans. CONCLUSIONS Multiple novel therapies are currently under active investigation to slow or stop noncompressible torso hemorrhage in the prehospital setting and show promising results.
Collapse
Affiliation(s)
- Mia K Klein
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nick D Tsihlis
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Timothy A Pritts
- Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Melina R Kibbe
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
24
|
Ben-Akiva E, Meyer RA, Yu H, Smith JT, Pardoll DM, Green JJ. Biomimetic anisotropic polymeric nanoparticles coated with red blood cell membranes for enhanced circulation and toxin removal. SCIENCE ADVANCES 2020; 6:eaay9035. [PMID: 32490199 PMCID: PMC7239698 DOI: 10.1126/sciadv.aay9035] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/22/2020] [Indexed: 05/17/2023]
Abstract
The design of next-generation nanobiomaterials requires precise engineering of both physical properties of the core material and chemical properties of the material's surface to meet a biological function. A bio-inspired modular and versatile technology was developed to allow biodegradable polymeric nanoparticles to circulate through the blood for extended periods of time while also acting as a detoxification device. To mimic red blood cells, physical and chemical biomimicry are combined to enhance the biological function of nanomaterials in vitro and in vivo. The anisotropic shape and membrane coating synergize to resist cellular uptake and reduce clearance from the blood. This approach enhances the detoxification properties of nanoparticles, markedly improving survival in a mouse model of sepsis. The anisotropic membrane-coated nanoparticles have enhanced biodistribution and therapeutic efficacy. These biomimetic biodegradable nanodevices and their derivatives have promise for applications ranging from detoxification agents, to drug delivery vehicles, and to biological sensors.
Collapse
Affiliation(s)
- Elana Ben-Akiva
- Departments of Biomedical Engineering, Materials Science and Engineering, Chemical and Biomolecular Engineering, and Oncology, Translational Tissue Engineering Center, Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N Broadway, Smith Building 5017, Baltimore, MD 21231, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Randall A. Meyer
- Departments of Biomedical Engineering, Materials Science and Engineering, Chemical and Biomolecular Engineering, and Oncology, Translational Tissue Engineering Center, Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N Broadway, Smith Building 5017, Baltimore, MD 21231, USA
| | - Hongzhe Yu
- Departments of Biomedical Engineering, Materials Science and Engineering, Chemical and Biomolecular Engineering, and Oncology, Translational Tissue Engineering Center, Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N Broadway, Smith Building 5017, Baltimore, MD 21231, USA
| | - Jonathan T. Smith
- Departments of Biomedical Engineering, Materials Science and Engineering, Chemical and Biomolecular Engineering, and Oncology, Translational Tissue Engineering Center, Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N Broadway, Smith Building 5017, Baltimore, MD 21231, USA
| | - Drew M. Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Departments of Oncology, Medicine, Pathology and Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jordan J. Green
- Departments of Biomedical Engineering, Materials Science and Engineering, Chemical and Biomolecular Engineering, and Oncology, Translational Tissue Engineering Center, Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N Broadway, Smith Building 5017, Baltimore, MD 21231, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Corresponding author.
| |
Collapse
|
25
|
Abstract
Hemorrhage is the leading cause of preventable death in combat trauma and the secondary cause of death in civilian trauma. A significant number of deaths due to hemorrhage occur before and in the first hour after hospital arrival. A literature search was performed through PubMed, Scopus, and Institute of Scientific Information databases for English language articles using terms relating to hemostatic agents, prehospital, battlefield or combat dressings, and prehospital hemostatic resuscitation, followed by cross-reference searching. Abstracts were screened to determine relevance and whether appropriate further review of the original articles was warranted. Based on these findings, this paper provides a review of a variety of hemostatic agents ranging from clinically approved products for human use to newly developed concepts with great potential for use in prehospital settings. These hemostatic agents can be administered either systemically or locally to stop bleeding through different mechanisms of action. Comparisons of current hemostatic products and further directions for prehospital hemorrhage control are also discussed.
Collapse
Affiliation(s)
- Henry T Peng
- Defence Research and Development Canada, Toronto Research Centre, 1133 Sheppard Avenue West, Toronto, ON, M3K 2C9, Canada.
| |
Collapse
|
26
|
Shen J, Nada AA, Abou-Zeid NY, Hudson SM. Synthesis of chitosan iodoacetamides via carbodiimide coupling reaction: Effect of degree of substitution on the hemostatic properties. Carbohydr Polym 2020; 229:115522. [DOI: 10.1016/j.carbpol.2019.115522] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 08/23/2019] [Accepted: 10/22/2019] [Indexed: 11/16/2022]
|
27
|
Wang X, Liu Q, Sui J, Ramakrishna S, Yu M, Zhou Y, Jiang X, Long Y. Recent Advances in Hemostasis at the Nanoscale. Adv Healthc Mater 2019; 8:e1900823. [PMID: 31697456 DOI: 10.1002/adhm.201900823] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/17/2019] [Indexed: 01/13/2023]
Abstract
Rapid and effective hemostatic materials have received wide attention not only in the battlefield but also in hospitals and clinics. Traditional hemostasis relies on materials with little designability which has many limitations. Nanohemostasis has been proposed since the use of peptides in hemostasis. Nanomaterials exhibit excellent adhesion, versatility, and designability compared to traditional materials, laying a good foundation for future hemostatic materials. This review first summarizes current hemostatic methods and materials, and then introduces several cutting-edge designs and applications of nanohemostatic materials such as polypeptide assembly, electrospinning of cyanoacrylate, and nanochitosan. Particularly, their advantages and working mechanisms are introduced. Finally, the challenges and prospects of nanohemostasis are discussed.
Collapse
Affiliation(s)
- Xiao‐Xiong Wang
- Collaborative Innovation Center for Nanomaterials & DevicesCollege of PhysicsQingdao University Qingdao 266071 China
| | - Qi Liu
- Collaborative Innovation Center for Nanomaterials & DevicesCollege of PhysicsQingdao University Qingdao 266071 China
| | - Jin‐Xia Sui
- Collaborative Innovation Center for Nanomaterials & DevicesCollege of PhysicsQingdao University Qingdao 266071 China
| | - Seeram Ramakrishna
- Collaborative Innovation Center for Nanomaterials & DevicesCollege of PhysicsQingdao University Qingdao 266071 China
- Center for Nanofibers & NanotechnologyNational University of Singapore Singapore 119077 Singapore
| | - Miao Yu
- Collaborative Innovation Center for Nanomaterials & DevicesCollege of PhysicsQingdao University Qingdao 266071 China
- Department of Mechanical EngineeringColumbia University New York NY 10027 USA
| | - Yu Zhou
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesQingdao University Qingdao 266071 China
| | - Xing‐Yu Jiang
- Laboratory for Biological Effects of Nanomaterials & NanosafetyNational Center for Nanoscience & Technology Beijing 100190 China
| | - Yun‐Ze Long
- Collaborative Innovation Center for Nanomaterials & DevicesCollege of PhysicsQingdao University Qingdao 266071 China
| |
Collapse
|
28
|
Improving the anticancer effect of afatinib and microRNA by using lipid polymeric nanoparticles conjugated with dual pH-responsive and targeting peptides. J Nanobiotechnology 2019; 17:89. [PMID: 31426807 PMCID: PMC6699136 DOI: 10.1186/s12951-019-0519-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
Background The emergence of resistance to chemotherapy or target therapy, tumor metastasis, and systemic toxicity caused by available anticancer drugs hamper the successful colorectal cancer (CRC) treatment. The rise in epidermal growth factor receptor (EGFR; human epidermal growth factor receptor 1; HER1) expression and enhanced phosphorylation of HER2 and HER3 are associated with tumor resistance, metastasis and invasion, thus resulting in poor outcome of anti-CRC therapy. The use of afatinib, a pan-HER inhibitor, is a potential therapeutic approach for resistant CRC. Additionally, miR-139 has been reported to be negatively correlated with chemoresistance, metastasis, and epithelial–mesenchymal transition (EMT) of CRC. Hence, we develop a nanoparticle formulation consisting of a polymer core to carry afatinib or miR-139, which is surrounded by lipids modified with a targeting ligand and a pH-sensitive penetrating peptide to improve the anticancer effect of cargos against CRC cells. Results Our findings show that this formulation displays a spherical shape with core/shell structure, homogeneous particle size distribution and negative zeta potential. The prepared formulations demonstrate a pH-sensitive release profile and an enhanced uptake of cargos into human colorectal adenocarcinoma Caco-2 cells in response to the acidic pH. This nanoparticle formulation incorporating afatinib and miR-139 exhibits low toxicity to normal cells but shows a better inhibitory effect on Caco-2 cells than other formulations. Moreover, the encapsulation of afatinib and miR-139 in peptide-modified nanoparticles remarkably induces apoptosis and inhibits migration and resistance of Caco-2 cells via suppression of pan-HER tyrosine kinase/multidrug resistance/metastasis pathways. Conclusion This study proposes a multifunctional nanoparticle formulation for targeted modulation of apoptosis/EGFR/HER/EMT/resistance/progression pathways to increase the sensitivity of colon cancer cells to afatinib. Electronic supplementary material The online version of this article (10.1186/s12951-019-0519-6) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Gkikas M, Peponis T, Mesar T, Hong C, Avery RK, Roussakis E, Yoo HJ, Parakh A, Patino M, Sahani DV, Watkins MT, Oklu R, Evans CL, Albadawi H, Velmahos G, Olsen BD. Systemically Administered Hemostatic Nanoparticles for Identification and Treatment of Internal Bleeding. ACS Biomater Sci Eng 2019; 5:2563-2576. [PMID: 33405762 DOI: 10.1021/acsbiomaterials.9b00054] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Internal bleeding is an injury that can be difficult to localize and effectively treat without invasive surgeries. Injectable polymeric nanoparticles have been developed that can reduce clotting times and blood loss, but they have yet to incorporate sufficient diagnostic capabilities to assist in identifying bleeding sources. Herein, polymeric nanoparticles were developed to simultaneously treat internal bleeding while incorporating tracers for visualization of the nanoparticles by standard clinical imaging modalities. Addition of 1,1'-dioctadecyl-3,3,3',3'-tetramethylindodicarbocyanine perchlorate (DiD; a fluorescent dye), biotin functionality, and gold nanoparticles to hemostatic polymeric nanoparticles resulted in nanoparticles amenable to imaging with near-infrared (NIR) imaging, immunohistochemistry, and X-ray computed tomography (CT), respectively. Following a lethal liver resection injury, visualization of accumulated nanoparticles by multiple imaging methods was achieved in rodents, with the highest accumulation observed at the liver injury site, resulting in improved survival rates. Tracer addition to therapeutic nanoparticles allows for an expansion of their applicability, during stabilization by first responders to diagnosis and identification of unknown internal bleeding sites by clinicians using standard clinical imaging modalities.
Collapse
Affiliation(s)
- Manos Gkikas
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Chemistry, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| | - Thomas Peponis
- Department of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02144, United States
| | - Tomaz Mesar
- Department of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02144, United States
| | - Celestine Hong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Reginald K Avery
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Emmanuel Roussakis
- Wellman Center for Photomedicine, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
| | - Hyung-Jin Yoo
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, Massachusetts 02144, United States
| | - Anushri Parakh
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02144, United States
| | - Manuel Patino
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02144, United States
| | - Dushyant V Sahani
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02144, United States
| | - Michael T Watkins
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, Massachusetts 02144, United States
| | - Rahmi Oklu
- Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Arizona 85259, United States
| | - Conor L Evans
- Wellman Center for Photomedicine, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
| | - Hassan Albadawi
- Division of Vascular and Interventional Radiology, Mayo Clinic, Scottsdale, Arizona 85259, United States
| | - George Velmahos
- Department of Trauma, Emergency Surgery and Surgical Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02144, United States
| | - Bradley D Olsen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
30
|
Askri D, Ouni S, Galai S, Chovelon B, Arnaud J, Sturm N, Lehmann SG, Sakly M, Amara S, Sève M. Nanoparticles in foods? A multiscale physiopathological investigation of iron oxide nanoparticle effects on rats after an acute oral exposure: Trace element biodistribution and cognitive capacities. Food Chem Toxicol 2019; 127:173-181. [DOI: 10.1016/j.fct.2019.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/06/2019] [Accepted: 03/10/2019] [Indexed: 12/16/2022]
|
31
|
He Y, Xu J, Sun X, Ren X, Maharjan A, York P, Su Y, Li H, Zhang J. Cuboidal tethered cyclodextrin frameworks tailored for hemostasis and injured vessel targeting. Am J Cancer Res 2019; 9:2489-2504. [PMID: 31131049 PMCID: PMC6525997 DOI: 10.7150/thno.31159] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022] Open
Abstract
Rationale: Targeted delivery of therapeutic drugs or imaging agents to injured blood vessels via nanocarriers is likely to be dependent on the particle shape, yet cubic nanoparticle carriers have not been reported for vascular targeting. Here, we demonstrate that cuboidal cyclodextrin frameworks possess superior hemostasis effect and injured vessels targeting compared with spherical counterpart. Methods: Cuboidal and biocompatible γ-cyclodextrin metal-organic frameworks (CD-MOFs) are synthesized, tethered via crosslinking and surface modification with GRGDS peptide (GS5-MOFs). The specific interactions of cubic GS5-MOF nanoparticles with activated platelets were investigated by in vitro platelet aggregation assay and atomic force microscopy measurements (AFM). The hemostatic capacity and injured vessel targeting efficacy were evaluated in vivo. Results: Cuboidal GS5-MOF nanoparticles exhibit enhanced adhesion and aggregation with activated platelets in vitro under static condition and a physiologically relevant flow environment. The cubic GS5-MOF nanoparticles show efficient hemostatic effects with bleeding time and blood loss decrease of 90% and strong injured vessel targeting in vivo, markedly superior to spherical γ-CD nanosponges with the same chemical composition. Conclusions: These results clearly highlight the contribution of the cuboidal shape of GS5-MOFs to the enhanced aggregation of activated platelets and high targeting to damaged vessels. The cuboidal nanoparticle system provides an innovative delivery platform for the treatment and diagnosis of vascular diseases.
Collapse
|
32
|
Intravenous administration of synthetic platelets (SynthoPlate) in a mouse liver injury model of uncontrolled hemorrhage improves hemostasis. J Trauma Acute Care Surg 2019. [PMID: 29538234 DOI: 10.1097/ta.0000000000001893] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Clinical resuscitative treatment of traumatic hemorrhage involves transfusion of RBC, platelets and plasma in controlled ratios. However, use of such blood components, especially platelets, present many challenges including availability, portability, contamination risks, and short shelf-life, which limit the use of platelet transfusions outside of large trauma centers such as remote civilian hospitals and austere prehospital settings. This has prompted significant research in platelet substitutes that may resolve the above issues while providing platelet-mimetic hemostatic action. In this framework, we have developed a synthetic platelet surrogate, SynthoPlate, by integrative decoration of platelet function mimetic peptides on a biocompatible lipid nanovesicle platform. We have previously demonstrated hemostatic capability of SynthoPlate in correcting tail-bleeding time in thrombocytopenic mice. Building on this, we hypothesized that SynthoPlate transfusion would decrease bleeding in a murine model of acute hemorrhagic shock. METHODS A validated model of uncontrolled intraperitoneal hemorrhage, via liver laceration was used to induce hemorrhagic shock in mice. SynthoPlate, control (unmodified) particles, and normal saline were administered as pretreatment and recue infusions to mice undergoing liver laceration and evaluated for hemostatic benefit by determining differences in blood loss and monitoring real-time hemodynamic data. RESULTS Pretreatment SynthoPlate transfusion resulted in significant reduction of blood loss following hemorrhage, compared with control particles or normal saline treatment (0.86 ± 0.16 g control particles [CP] vs. 0.84 ± 0.13 g normal saline [NS] vs. 0.68 ± 0.09 g SynthoPlate, p < 0.005). SynthoPlate transfused mice demonstrated improved hemodynamics taking significantly longer to develop post-injury hypotension (168.3 ± 106.6 seconds CP vs. 137 ± 58 seconds NS vs. 546.7 ± 329.8 seconds SynthoPlate, p < 0.05). SynthoPlate infusion following liver laceration, that is, rescue transfusion, also resulted in a significant decrease in blood loss (0.89 ± 0.17 g CP vs. 0.92 ± 0.19 g NS vs. 0.69 ± 0.18 g SynthoPlate, p < 0.05). CONCLUSION Transfusion of SynthoPlate particles reduces blood loss in a murine model of liver injury, and SynthoPlates may represent a viable transfusion product for the mitigation of blood loss in acute, severe hemorrhagic shock.
Collapse
|
33
|
Zhang P, Li S, Zhang S, Zhang X, Wan L, Yun Z, Ji S, Gong F, Huang M, Wang L, Zhu X, Tan Y, Wan Y. GRGDS-functionalized chitosan nanoparticles as a potential intravenous hemostat for traumatic hemorrhage control in an animal model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2531-2540. [PMID: 30193814 DOI: 10.1016/j.nano.2018.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/15/2018] [Accepted: 08/08/2018] [Indexed: 01/25/2023]
Abstract
Hemostats, which are used for immediate intervention during internal hemorrhage in order to reduce resulting mortality and morbidity, are relatively rare. Here, we describe novel intravenous nanoparticles (CPG-NPs-2000) with chitosan succinate (CSS) as cores, polyethylene glycol (PEG-2000) as spacers and a glycine-arginine-glycine-aspartic acid-serine (GRGDS) peptide as targeted, active hemostatic motifs. CPG-NPs-2000 displayed significant hemostatic efficacy, compared to the saline control, CSS nanoparticles, and tranexamic acid in liver trauma rat models. Further studies have demonstrated that CPG-NPs-2000 are effectively cleared from organs and blood, within 2 and 48 h, respectively. In addition, administration of CPG-NPs-2000 does not affect clotting function under normal physiological conditions, indicating their potential safety in vivo. CPG-NPs-2000 exhibit excellent thermal stability, good solubility, and redistribution ability, in addition to being low cost. These characteristics indicate that CPG-NPs-2000 may have strong potential as effective intravenous hemostats for treating severe internal bleeding.
Collapse
Affiliation(s)
- Pingyi Zhang
- School of Chemical Engineering and Technology, Guangdong Engineering Technology Research Center for Platform Chemicals from Marine Biomass and Their Functionalization, Sun Yat-sen University, Guangzhou, China
| | - Subo Li
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Shikun Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Xue Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Luming Wan
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Zhimin Yun
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Shouping Ji
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Feng Gong
- Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Manna Huang
- School of Chemical Engineering and Technology, Guangdong Engineering Technology Research Center for Platform Chemicals from Marine Biomass and Their Functionalization, Sun Yat-sen University, Guangzhou, China
| | - Leilei Wang
- School of Chemical Engineering and Technology, Guangdong Engineering Technology Research Center for Platform Chemicals from Marine Biomass and Their Functionalization, Sun Yat-sen University, Guangzhou, China
| | - Xinhai Zhu
- School of Chemical Engineering and Technology, Guangdong Engineering Technology Research Center for Platform Chemicals from Marine Biomass and Their Functionalization, Sun Yat-sen University, Guangzhou, China
| | - Yingxia Tan
- Institute of Health Service and Transfusion Medicine, Beijing, China.
| | - Yiqian Wan
- School of Chemical Engineering and Technology, Guangdong Engineering Technology Research Center for Platform Chemicals from Marine Biomass and Their Functionalization, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
34
|
Humbrecht C, Kientz D, Gachet C. Platelet transfusion: Current challenges. Transfus Clin Biol 2018; 25:151-164. [PMID: 30037501 DOI: 10.1016/j.tracli.2018.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 12/29/2022]
Abstract
Since the late sixties, platelet concentrates are transfused to patients presenting with severe thrombocytopenia, platelet function defects, injuries, or undergoing surgery, to prevent the risk of bleeding or to treat actual hemorrhage. Current practices differ according to the country or even in different hospitals and teams. Although crucial advances have been made during the last decades, questions and debates still arise about the right doses to transfuse, the use of prophylactic or therapeutic strategies, the nature and quality of PC, the storage conditions, the monitoring of transfusion efficacy and the microbiological and immunological safety of platelet transfusion. Finally, new challenges are emerging with potential new platelet products, including cold stored or in vitro produced platelets. The most debated of these points are reviewed.
Collapse
Affiliation(s)
- C Humbrecht
- Établissement français du sang grand est, 85-87, boulevard Lobau, 54064 Nancy cedex, France.
| | - D Kientz
- Établissement français du sang grand est, 85-87, boulevard Lobau, 54064 Nancy cedex, France
| | - C Gachet
- Établissement français du sang grand est, 85-87, boulevard Lobau, 54064 Nancy cedex, France.
| |
Collapse
|
35
|
Hemostatic nanoparticles increase survival, mitigate neuropathology and alleviate anxiety in a rodent blast trauma model. Sci Rep 2018; 8:10622. [PMID: 30006635 PMCID: PMC6045585 DOI: 10.1038/s41598-018-28848-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 06/21/2018] [Indexed: 12/22/2022] Open
Abstract
Explosions account for 79% of combat related injuries and often lead to polytrauma, a majority of which include blast-induced traumatic brain injuries (bTBI). These injuries lead to internal bleeding in multiple organs and, in the case of bTBI, long term neurological deficits. Currently, there are no treatments for internal bleeding beyond fluid resuscitation and surgery. There is also a dearth of treatments for TBI. We have developed a novel approach using hemostatic nanoparticles that encapsulate an anti-inflammatory, dexamethasone, to stop the bleeding and reduce inflammation after injury. We hypothesize that this will improve not only survival but long term functional outcomes after blast polytrauma. Poly(lactic-co-glycolic acid) hemostatic nanoparticles encapsulating dexamethasone (hDNPs) were fabricated and tested following injury along with appropriate controls. Rats were exposed to a single blast wave using an Advanced Blast Simulator, inducing primary blast lung and bTBI. Survival was elevated in the hDNPs group compared to controls. Elevated anxiety parameters were found in the controls, compared to hDNPs. Histological analysis indicated that apoptosis and blood-brain barrier disruption in the amygdala were significantly increased in the controls compared to the hDNPs and sham groups. Immediate intervention is crucial to mitigate injury mechanisms that contribute to emotional deficits.
Collapse
|
36
|
Onwukwe C, Maisha N, Holland M, Varley M, Groynom R, Hickman D, Uppal N, Shoffstall A, Ustin J, Lavik E. Engineering Intravenously Administered Nanoparticles to Reduce Infusion Reaction and Stop Bleeding in a Large Animal Model of Trauma. Bioconjug Chem 2018; 29:2436-2447. [PMID: 29965731 DOI: 10.1021/acs.bioconjchem.8b00335] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bleeding from traumatic injury is the leading cause of death for young people across the world, but interventions are lacking. While many agents have shown promise in small animal models, translating the work to large animal models has been exceptionally difficult in great part because of infusion-associated complement activation to nanomaterials that leads to cardiopulmonary complications. Unfortunately, this reaction is seen in at least 10% of the population. We developed intravenously infusible hemostatic nanoparticles that were effective in stopping bleeding and improving survival in rodent models of trauma. To translate this work, we developed a porcine liver injury model. Infusion of the first generation of hemostatic nanoparticles and controls 5 min after injury led to massive vasodilation and exsanguination even at extremely low doses. In naïve animals, the physiological changes were consistent with a complement-associated infusion reaction. By tailoring the zeta potential, we were able to engineer a second generation of hemostatic nanoparticles and controls that did not exhibit the complement response at low and moderate doses but did at the highest doses. These second-generation nanoparticles led to cessation of bleeding within 10 min of administration even though some signs of vasodilation were still seen. While the complement response is still a challenge, this work is extremely encouraging in that it demonstrates that when the infusion-associated complement response is managed, hemostatic nanoparticles are capable of rapidly stopping bleeding in a large animal model of trauma.
Collapse
Affiliation(s)
- Chimdiya Onwukwe
- University of Maryland Baltimore County , 1000 Hilltop Circle, Baltimore , Maryland 21050 , United States
| | - Nuzhat Maisha
- University of Maryland Baltimore County , 1000 Hilltop Circle, Baltimore , Maryland 21050 , United States
| | - Mark Holland
- University of Maryland Baltimore County , 1000 Hilltop Circle, Baltimore , Maryland 21050 , United States
| | - Matt Varley
- Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Rebecca Groynom
- Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - DaShawn Hickman
- Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Nishant Uppal
- Harvard Medical School , 25 Shattuck Street , Boston , Massachusetts 02115 , United States
| | - Andrew Shoffstall
- Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Jeffrey Ustin
- Case Western Reserve University , 10900 Euclid Avenue , Cleveland , Ohio 44106 , United States
| | - Erin Lavik
- University of Maryland Baltimore County , 1000 Hilltop Circle, Baltimore , Maryland 21050 , United States
| |
Collapse
|
37
|
Fernandez-Moure J, Maisha N, Lavik EB, Cannon JW. The Chemistry of Lyophilized Blood Products. Bioconjug Chem 2018; 29:2150-2160. [PMID: 29791137 DOI: 10.1021/acs.bioconjchem.8b00271] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
With the development of new biologics and bioconjugates, storage and preservation have become more critical than ever before. Lyophilization is a method of cell and protein preservation by removing a solvent such as water from a substance followed by freezing. This technique has been used in the past and still holds promise for overcoming logistic challenges in safety net hospitals with limited blood banking resources, austere environments such as combat, and mass casualty situations where existing resources may be outstripped. This method allows for long-term storage and transport but requires the bioconjugation of preservatives to prevent cell destabilization. Trehalose is utilized as a bioconjugate in platelet and red blood cell preservation to maintain protein thermodynamics and stabilizing protein formulations in liquid and freeze-dried states. Biomimetic approaches have been explored as alternatives to cryo- and lyopreservation of blood components. Intravascular hemostats such as PLGA nanoparticles functionalized with PEG motifs, topical hemostats utilizing fibrinogen or chitosan, and liposomal encapsulated hemoglobin with surface modifications are effectively stored long-term through bioconjugation. In thinking about the best methods for storage and transport, we are focusing this topical review on blood products that have the longest track record of preservation and looking at how these methods can be applied to synthetic systems.
Collapse
Affiliation(s)
- Joseph Fernandez-Moure
- Division of Trauma, Surgical Critical Care & Emergency Surgery , Perelman School of Medicine at the University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Nuzhat Maisha
- Department of Chemical, Biochemical & Environmental Engineering , University of Maryland, Baltimore County , Baltimore , Maryland 21250 , United States
| | - Erin B Lavik
- Department of Chemical, Biochemical & Environmental Engineering , University of Maryland, Baltimore County , Baltimore , Maryland 21250 , United States
| | - Jeremy W Cannon
- Division of Trauma, Surgical Critical Care & Emergency Surgery , Perelman School of Medicine at the University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States.,Department of Surgery , Uniformed Services University of the Health Sciences , Bethesda , Maryland 20814 , United States
| |
Collapse
|
38
|
Welsch N, Brown AC, Barker TH, Lyon LA. Enhancing clot properties through fibrin-specific self-cross-linked PEG side-chain microgels. Colloids Surf B Biointerfaces 2018; 166:89-97. [PMID: 29549720 PMCID: PMC6050065 DOI: 10.1016/j.colsurfb.2018.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/14/2018] [Accepted: 03/01/2018] [Indexed: 02/06/2023]
Abstract
Excessive bleeding and resulting complications are a major cause of death in both trauma and surgical settings. Recently, there have been a number of investigations into the design of synthetic hemostatic agents with platelet-mimicking activity to effectively treat patients suffering from severe hemorrhage. We developed platelet-like particles from microgels composed of polymers carrying polyethylene glycol (PEG) side-chains and fibrin-targeting single domain variable fragment antibodies (PEG-PLPs). Comparable to natural platelets, PEG-PLPs were found to enhance the fibrin network formation in vitro through strong adhesion to the emerging fibrin clot and physical, non-covalent cross-linking of nascent fibrin fibers. Furthermore, the mechanical reinforcement of the fibrin mesh through the incorporation of particles into the network leads to a ∼three-fold decrease of the overall clot permeability as compared to control clots. However, transport of biomolecules through the fibrin clots, such as peptides and larger proteins is not hindered by the presence of PEG-PLPs and the altered microstructure. Compared to control clots with an elastic modulus of 460+/-260 Pa, PEG-PLP-reinforced fibrin clots exhibit higher degrees of stiffness as demonstrated by the significantly increased average Younǵs modulus of 1770 +/±720 Pa, as measured by AFM force spectroscopy. Furthermore, in vitro degradation studies with plasmin demonstrate that fibrin clots formed in presence of PEG-PLPs withstand hydrolysis for 24 h, indicating enhanced stabilization against exogenous fibrinolysis. The entire set of data suggests that the designed platelet-like particles have high potential for use as hemostatic agents in emergency medicine and surgical settings.
Collapse
Affiliation(s)
- Nicole Welsch
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ashley C Brown
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA; Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina, Chapel Hill, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Thomas H Barker
- The Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - L Andrew Lyon
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA; Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA.
| |
Collapse
|
39
|
Hickman DA, Pawlowski CL, Sekhon UDS, Marks J, Gupta AS. Biomaterials and Advanced Technologies for Hemostatic Management of Bleeding. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:10.1002/adma.201700859. [PMID: 29164804 PMCID: PMC5831165 DOI: 10.1002/adma.201700859] [Citation(s) in RCA: 331] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 06/18/2017] [Indexed: 05/03/2023]
Abstract
Bleeding complications arising from trauma, surgery, and as congenital, disease-associated, or drug-induced blood disorders can cause significant morbidities and mortalities in civilian and military populations. Therefore, stoppage of bleeding (hemostasis) is of paramount clinical significance in prophylactic, surgical, and emergency scenarios. For externally accessible injuries, a variety of natural and synthetic biomaterials have undergone robust research, leading to hemostatic technologies including glues, bandages, tamponades, tourniquets, dressings, and procoagulant powders. In contrast, treatment of internal noncompressible hemorrhage still heavily depends on transfusion of whole blood or blood's hemostatic components (platelets, fibrinogen, and coagulation factors). Transfusion of platelets poses significant challenges of limited availability, high cost, contamination risks, short shelf-life, low portability, performance variability, and immunological side effects, while use of fibrinogen or coagulation factors provides only partial mechanisms for hemostasis. With such considerations, significant interdisciplinary research endeavors have been focused on developing materials and technologies that can be manufactured conveniently, sterilized to minimize contamination and enhance shelf-life, and administered intravenously to mimic, leverage, and amplify physiological hemostatic mechanisms. Here, a comprehensive review regarding the various topical, intracavitary, and intravenous hemostatic technologies in terms of materials, mechanisms, and state-of-art is provided, and challenges and opportunities to help advancement of the field are discussed.
Collapse
Affiliation(s)
- DaShawn A Hickman
- Case Western Reserve University School of Medicine, Department of Pathology, Cleveland, Ohio 44106, USA
| | - Christa L Pawlowski
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, Ohio 44106, USA
| | - Ujjal D S Sekhon
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, Ohio 44106, USA
| | - Joyann Marks
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, Ohio 44106, USA
| | - Anirban Sen Gupta
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, Ohio 44106, USA
| |
Collapse
|
40
|
Hangge P, Stone J, Albadawi H, Zhang YS, Khademhosseini A, Oklu R. Hemostasis and nanotechnology. Cardiovasc Diagn Ther 2017; 7:S267-S275. [PMID: 29399530 DOI: 10.21037/cdt.2017.08.07] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hemorrhage accounts for significant morbidity and mortality. Various techniques have been employed to augment hemostasis from simple tourniquets to self-assembling nanoparticles. A growing understanding of the natural clotting cascade has allowed agents to become more targeted for potential use in different clinical scenarios. This review discusses current and developing hemostatic techniques, including matrix agents, external agents, biologically inspired agents, and synthetic and cell-derived nanoparticles.
Collapse
Affiliation(s)
- Patrick Hangge
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Jonathan Stone
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Hassan Albadawi
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| | - Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Rahmi Oklu
- Division of Interventional Radiology, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
41
|
Gaston E, Fraser JF, Xu ZP, Ta HT. Nano- and micro-materials in the treatment of internal bleeding and uncontrolled hemorrhage. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:507-519. [PMID: 29162534 DOI: 10.1016/j.nano.2017.11.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/01/2017] [Accepted: 11/05/2017] [Indexed: 12/14/2022]
Abstract
Internal bleeding is defined as the loss of blood that occurs inside of a body cavity. After a traumatic injury, hemorrhage accounts for over 35% of pre-hospital deaths and 40% of deaths within the first 24 hours. Coagulopathy, a disorder in which the blood is not able to properly form clots, typically develops after traumatic injury and results in a higher rate of mortality. The current methods to treat internal bleeding and coagulopathy are inadequate due to the requirement of extensive medical equipment that is typically not available at the site of injury. To discover a potential route for future research, several current and novel treatment methods have been reviewed and analyzed. The aim of investigating different potential treatment options is to expand available knowledge, while also call attention to the importance of research in the field of treatment for internal bleeding and hemorrhage due to trauma.
Collapse
Affiliation(s)
- Elizabeth Gaston
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD, Australia; Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - John F Fraser
- Faculty of Medicine, Critical Care Research Group, Prince Charles Hospital and the University of Queensland, Brisbane, Brisbane, QLD, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD, Australia
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
42
|
Sen Gupta A. Bio-inspired nanomedicine strategies for artificial blood components. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:10.1002/wnan.1464. [PMID: 28296287 PMCID: PMC5599317 DOI: 10.1002/wnan.1464] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/23/2017] [Accepted: 01/29/2017] [Indexed: 11/12/2022]
Abstract
Blood is a fluid connective tissue where living cells are suspended in noncellular liquid matrix. The cellular components of blood render gas exchange (RBCs), immune surveillance (WBCs) and hemostatic responses (platelets), and the noncellular components (salts, proteins, etc.) provide nutrition to various tissues in the body. Dysfunction and deficiencies in these blood components can lead to significant tissue morbidity and mortality. Consequently, transfusion of whole blood or its components is a clinical mainstay in the management of trauma, surgery, myelosuppression, and congenital blood disorders. However, donor-derived blood products suffer from issues of shortage in supply, need for type matching, high risks of pathogenic contamination, limited portability and shelf-life, and a variety of side-effects. While robust research is being directed to resolve these issues, a parallel clinical interest has developed toward bioengineering of synthetic blood substitutes that can provide blood's functions while circumventing the above problems. Nanotechnology has provided exciting approaches to achieve this, using materials engineering strategies to create synthetic and semi-synthetic RBC substitutes for enabling oxygen transport, platelet substitutes for enabling hemostasis, and WBC substitutes for enabling cell-specific immune response. Some of these approaches have further extended the application of blood cell-inspired synthetic and semi-synthetic constructs for targeted drug delivery and nanomedicine. The current study provides a comprehensive review of the various nanotechnology approaches to design synthetic blood cells, along with a critical discussion of successes and challenges of the current state-of-art in this field. WIREs Nanomed Nanobiotechnol 2017, 9:e1464. doi: 10.1002/wnan.1464 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
43
|
Fibrinogen γ-Chain Peptide-Coated Adenosine 5' Diphosphate-Encapsulated Liposomes Rescue Mice From Lethal Blast Lung Injury via Adenosine Signaling. Crit Care Med 2017; 44:e827-37. [PMID: 27054893 DOI: 10.1097/ccm.0000000000001707] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVES Fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes can accumulate via dodecapeptide HHLGGAKQAGDV interactions at bleeding sites where they release adenosine 5'-diphosphate that is rapidly metabolized to adenosine, which has tissue-protective effects. We investigated the efficacy of fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes to treat blast lung injury, with a focus on adenosine signaling. DESIGN Controlled animal study. SETTING University research laboratory. SUBJECTS Adult male C57BL/6 mice. INTERVENTIONS Mice were pretreated with fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes, dodecapeptide HHLGGAKQAGDV-(phosphate-buffered saline)-liposomes, adenosine 5' diphosphateliposomes, or phosphate-buffered saline-liposomes. Five minutes after treatment the mice received a single laser-induced shock wave (1.8 J/cm) that caused lethal blast lung injury, and their survival times and lung injuries were then assessed. We also evaluated the therapeutic effect of posttreatment with fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes or H12-(phosphate-buffered saline)-liposomes 1 minute after laser-induced shock wave exposure. To examine the effect of adenosine signaling, adenosine A2A receptor (ZM241385) or adenosine A2B receptor (PSB 1115) antagonists were administered to the mice 1 hour before the pretreatment with fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes that was followed by laser-induced shock wave exposure. MEASUREMENTS AND MAIN RESULTS Pre- and posttreatment with fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes significantly increased mouse survival [fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes: 58% survival vs H12-(phosphate-buffered saline)-liposomes: 8%; p < 0.05 (posttreatment)] and mitigated pulmonary tissue damage/hemorrhage and neutrophil accumulation after laser-induced shock wave exposure. fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes accumulated at pulmonary vessel injury sites after laser-induced shock wave exposure with both pre- and posttreatment. Furthermore, pretreatment with fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes reduced albumin and macrophage inflammatory protein-2 levels in bronchoalveolar lavage fluid. Although fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV)-coated adenosine 5'-diphosphate-encapsulated liposomes pretreatment did not affect blood coagulation activity in the injured mice, its beneficial effect on blast lung injury was significantly abrogated by A2A or A2B adenosine receptor antagonists (A2A antagonist: 17% survival; A2B antagonist: 33% vs dimethyl sulfoxide control: 80%; p < 0.05, respectively). CONCLUSIONS Fibrinogen γ-chain (dodecapeptide HHLGGAKQA GDV)-coated adenosine 5'-diphosphate-encapsulated liposomes may be effective against blast lung injury by promoting tissue-protective adenosine signaling and could represent a novel controlled-release drug delivery system.
Collapse
|
44
|
|
45
|
Green JJ, Elisseeff JH. Mimicking biological functionality with polymers for biomedical applications. Nature 2017; 540:386-394. [PMID: 27974772 DOI: 10.1038/nature21005] [Citation(s) in RCA: 319] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 09/12/2016] [Indexed: 12/12/2022]
Abstract
The vast opportunities for biomaterials design and functionality enabled by mimicking nature continue to stretch the limits of imagination. As both biological understanding and engineering capabilities develop, more sophisticated biomedical materials can be synthesized that have multifaceted chemical, biological and physical characteristics designed to achieve specific therapeutic goals. Mimicry is being used in the design of polymers for biomedical applications that are required locally in tissues, systemically throughout the body, and at the interface with tissues.
Collapse
Affiliation(s)
- Jordan J Green
- Translational Tissue Engineering Center, Departments of Biomedical Engineering and Ophthalmology, and the Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Departments of Biomedical Engineering and Ophthalmology, and the Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
46
|
Cheng J, Feng S, Han S, Zhang X, Chen Y, Zhou X, Wang R, Li X, Hu H, Zhang J. Facile Assembly of Cost-Effective and Locally Applicable or Injectable Nanohemostats for Hemorrhage Control. ACS NANO 2016; 10:9957-9973. [PMID: 27736084 DOI: 10.1021/acsnano.6b04124] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Currently, there is still unmet demand for effective and safe hemostats to control abnormal bleeding in different conditions. With the aim to develop affordable, safe, effective, easily stored, and low-cost hemostats, we developed a series of positively charged nanoparticles by a facile one-pot assembly approach. In this strategy, nanoparticles were formed by cholic-acid-mediated self-assembly of polyethylenimine (PEI). Regardless of different structures of cholic acids and PEIs, well-defined nanoparticles could be successfully formed. The assembly process was dominated by multiple interactions between cholic acid and PEI, including electrostatic, hydrogen bonding, and hydrophobic forces. In vitro studies showed that assembled nanoparticles effectively induced aggregation and activation of platelets. Local application of aqueous solution containing nanoparticles assembled by different cholic acids and PEIs significantly reduced bleeding times in different rodent models including tail transection in mice as well as liver bleeding and femoral artery bleeding in rats or rabbits. Moreover, intravenous (i.v.) injection of this type of positively charged nanoparticles notably prevented bleeding in the femoral artery in rats by targeting the injured site via opsonization of nanoparticles with fibrinogen. By contrast, a control negatively charged nanoparticle showed no hemostatic activity after i.v. delivery. Also, preliminary evaluations in rats revealed a good safety profile after i.v. administration of assembled nanoparticles at a dose 4-fold higher than that used for hemostasis. These results demonstrated that cholic acid/PEI-assembled positive nanoparticles may function as cost-effective and locally applicable or injectable nanohemostats for hemorrhage control in the civilian setting and on the battlefield.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Taipa, Macau, China
| | | | | | | |
Collapse
|
47
|
Desborough MJR, Smethurst PA, Estcourt LJ, Stanworth SJ. Alternatives to allogeneic platelet transfusion. Br J Haematol 2016; 175:381-392. [PMID: 27650431 DOI: 10.1111/bjh.14338] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Allogeneic platelet transfusions are widely used for the prevention and treatment of bleeding in thrombocytopenia. Recent evidence suggests platelet transfusions have limited efficacy and are associated with uncertain immunomodulatory risks and concerns about viral or bacterial transmission. Alternatives to transfusion are a well-recognised tenet of Patient Blood Management, but there has been less focus on different strategies to reduce bleeding risk by comparison to platelet transfusion. Direct alternatives to platelet transfusion include agents to stimulate endogenous platelet production (thrombopoietin mimetics), optimising platelet adhesion to endothelium by treating anaemia or increasing von Willebrand factor levels (desmopressin), increasing formation of cross-linked fibrinogen (activated recombinant factor VII, fibrinogen concentrate or recombinant factor XIII), decreasing fibrinolysis (tranexamic acid or epsilon aminocaproic acid) or using artificial or modified platelets (cryopreserved platelets, lyophilised platelets, haemostatic particles, liposomes, engineered nanoparticles or infusible platelet membranes). The evidence base to support the use of these alternatives is variable, but an area of active research. Much of the current randomised controlled trial focus is on evaluation of the use of thrombopoietin mimetics and anti-fibrinolytics. It is also recognised that one alternative strategy to platelet transfusion is choosing not to transfuse at all.
Collapse
Affiliation(s)
- Michael J R Desborough
- NHS Blood and Transplant, John Radcliffe Hospital, Oxford, UK. .,Oxford Clinical Research in Transfusion Medicine, Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK.
| | | | - Lise J Estcourt
- NHS Blood and Transplant, John Radcliffe Hospital, Oxford, UK.,Oxford Clinical Research in Transfusion Medicine, Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| | - Simon J Stanworth
- NHS Blood and Transplant, John Radcliffe Hospital, Oxford, UK.,Oxford Clinical Research in Transfusion Medicine, Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
48
|
Abstract
Extremity injury is a significant burden to those injured in explosive incidents and local ischaemia can result in poor functionality in salvaged limbs. This study examined whether blast injury to a limb resulted in a change in endothelial phenotype leading to changes to the surrounding tissue. The hind limbs of terminally anaesthetized rabbits were subjected to one of four blast exposures (high, medium, low, or no blast). Blood samples were analyzed for circulating endothelial cells pre-injury and at 1, 6, and 11 h postinjury as well as analysis for endothelial activation pre-injury and at 1, 6, and 12 h postinjury. Post-mortem tissue (12 h post-injury) was analysed for both protein and mRNA expression and also for histopathology. The high blast group had significantly elevated levels of circulating endothelial cells 6 h postinjury. This group also had significantly elevated tissue mRNA expression of IL-6, E-selectin, TNF-α, HIF-1, thrombomodulin, and PDGF. There was a significant correlation between blast dose and the degree of tissue pathology (hemorrhage, neutrophil infiltrate, and oedema) with the worst scores in the high blast group. This study has demonstrated that blast injury can activate the endothelium and in some cases cause damage that in turn leads to pathological changes in the surrounding tissue. For the casualty injured by an explosion the damaging effects of hemorrhage and shock could be exacerbated by blast injury and vice versa so that even low levels of blast become damaging, all of which could affect tissue functionality and long-term outcomes.
Collapse
|
49
|
Abstract
Approaches to locally deliver drugs to specific regions of the body are being developed for many clinical applications, including treating hemorrhage. Increasing the concentration of therapeutic coagulants in areas where clots are forming and growing can be achieved by directing them to the injury, such as with catheters or external delivery devices, or by systemically administering therapeutics that target molecular signals of vascular damage. Treating severe hemorrhage by external measures is challenging because blood flow pushes hemostatic agents outward, reducing their efficacy. This review explains that self-propelling particles may be used for delivering therapeutics, such as coagulation factors, small molecules, or other chemical or biological agents, deep into wounds during hemorrhage. A recent example of self-propelling particles is highlighted, where propulsion enhanced the efficacy of a formulation of thrombin and tranexamic acid in treating bleeding in two murine models of hemorrhage and a porcine model of fatal, non-compressible hemorrhage. Many agents exist which modulate clotting, and novel approaches that facilitate their safe delivery to sites of vascular injury could reduce the enormous number of deaths from hemorrhage that occur globally.
Collapse
Affiliation(s)
- James R Baylis
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Karen Y T Chan
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Christian J Kastrup
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada.
| |
Collapse
|
50
|
Myerson JW, Anselmo AC, Liu Y, Mitragotri S, Eckmann DM, Muzykantov VR. Non-affinity factors modulating vascular targeting of nano- and microcarriers. Adv Drug Deliv Rev 2016; 99:97-112. [PMID: 26596696 PMCID: PMC4798918 DOI: 10.1016/j.addr.2015.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/29/2015] [Accepted: 10/09/2015] [Indexed: 12/22/2022]
Abstract
Particles capable of homing and adhering to specific vascular biomarkers have potential as fundamental tools in drug delivery for mediation of a wide variety of pathologies, including inflammation, thrombosis, and pulmonary disorders. The presentation of affinity ligands on the surface of a particle provides a means of targeting the particle to sites of therapeutic interest, but a host of other factors come into play in determining the targeting capacity of the particle. This review presents a summary of several key considerations in nano- and microparticle design that modulate targeted delivery without directly altering epitope-specific affinity. Namely, we describe the effect of factors in definition of the base carrier (including shape, size, and flexibility) on the capacity of carriers to access, adhere to, and integrate in target biological milieus. Furthermore, we present a summary of fundamental dynamics of carrier behavior in circulation, taking into account interactions with cells in circulation and the role of hemodynamics in mediating the direction of carriers to target sites. Finally, we note non-affinity aspects to uptake and intracellular trafficking of carriers in target cells. In total, recent findings presented here may offer an opportunity to capitalize on mitigating factors in the behavior of ligand-targeted carriers in order to optimize targeting.
Collapse
|