1
|
Lee SY, Choi JW, Hwang C, Park J, Karmakar M, Huh JW, Kim DD, Kim HC, Cho HJ. Intravascular Casting Radiopaque Hydrogel Systems for Transarterial Chemo/Cascade Catalytic/Embolization Therapy of Hepatocellular Carcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400287. [PMID: 39109954 DOI: 10.1002/smll.202400287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/27/2024] [Indexed: 11/21/2024]
Abstract
This paper introduces catheter-directed intravascular casting hydrogels for transarterial chemo/starvation/chemodynamic embolization (TACSCE) therapy of hepatocellular carcinoma (HCC). Comprising Mn ion-crosslinked hyaluronic acid-dopamine (HD) with glucose oxidase (for glucose decomposition to H2O2 in starvation therapy), doxorubicin (for chemotherapy), and iopamidol (for X-ray imaging), these hydrogels are fabricated for transarterial embolization therapy guided by X-ray fluoroscopy. Mn4+ (from MnO2) demonstrates strong coordination with the catechol group of HD, providing hypoxia relief through O2 generation and cellular glutathione (GSH) consumption, compared to the OH radical generation potential of Mn2+. The gelation time-controlled, catheter-injectable, and rheologically tuned multitherapeutic/embolic gel system effectively reaches distal arterioles, ensuring complete intravascular casting with fewer complications related to organic solvents. Glucose deprivation, cascade reactive oxygen species (ROS) generation, GSH depletion, and sustained release profiles of multiple drug cargos from the hydrogel system are also achieved. The combined chemo/starvation/chemodynamic efficacies of these designed hydrogel systems are confirmed in HCC cell cultures and HCC-bearing animal models. The developed radiopaque/injectable/embolic/sol-to-gel transformable systems for TACSCE therapy may offer enhanced therapeutic efficacies compared to typical transarterial embolization and transarterial chemoembolization procedures for HCC.
Collapse
Affiliation(s)
- Song Yi Lee
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jin Woo Choi
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - ChaeRim Hwang
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - JiHye Park
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Mrinmoy Karmakar
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ji Won Huh
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyo-Cheol Kim
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hyun-Jong Cho
- Department of Pharmacy, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
2
|
Wang X, Yuan K, Su Y, Li X, Meng L, Zhao N, Hu Y, Duan F, Xu FJ. Tuning Blood-Material Interactions to Generate Versatile Hemostatic Powders and Gels. Adv Healthc Mater 2024; 13:e2301945. [PMID: 37897223 DOI: 10.1002/adhm.202301945] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/05/2023] [Indexed: 10/29/2023]
Abstract
Polymer-based hemostatic materials/devices have been increasingly exploited for versatile clinical scenarios, while there is an urgent need to reveal the rational design/facile approach for procoagulant surfaces through regulating blood-material interactions. In this work, degradable powders (PLPS) and thermoresponsive gels (F127-PLPS) are readily developed as promising hemostatic materials for versatile clinical applications, through tuning blood-material interactions with optimized grafting of cationic polylysine: the former is facilely prepared by conjugating polylysine onto porous starch particle, while F127-PLPS is prepared by the simple mixture of PLPS and commercial thermosensitive polymer. In vitro and in vivo results demonstrate that PLPS2 with the optimal-/medium content of polylysine grafts achieve the superior hemostatic performance. The underlying procoagulant mechanism of PLPS2 surface is revealed as the selective fibrinogen adsorption among the competitive plasma-protein-adsorption process, which is the foundation of other blood-material interactions. Moreover, in vitro results confirm the achieved procoagulant surface of F127-PLPS through optimal PLPS2 loading. Together with the tunable thermoresponsiveness, F127-PLPS exhibits outstanding hemostatic utilization in both femoral-artery-injury and renal-artery-embolization models. The work thereby pioneers an appealing approach for generating versatile polymer-based hemostatic materials/devices.
Collapse
Affiliation(s)
- Xueru Wang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Kai Yuan
- Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Su
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoyue Li
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Limin Meng
- Department of Medical Imaging, Air Force Medical Center, PLA, Beijing, 100142, China
| | - Nana Zhao
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yang Hu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Feng Duan
- Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Material, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
3
|
Yathavan B, Chhibber T, Steinhauff D, Pulsipher A, Alt JA, Ghandehari H, Jafari P. Matrix-Mediated Delivery of Silver Nanoparticles for Prevention of Staphylococcus aureus and Pseudomonas aeruginosa Biofilm Formation in Chronic Rhinosinusitis. Pharmaceutics 2023; 15:2426. [PMID: 37896186 PMCID: PMC10610389 DOI: 10.3390/pharmaceutics15102426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023] Open
Abstract
Chronic rhinosinusitis (CRS) is a chronic health condition affecting the sinonasal cavity. CRS-associated mucosal inflammation leads to sinonasal epithelial cell death and epithelial cell barrier disruption, which may result in recurrent bacterial infections and biofilm formation. For patients who fail medical management and elect endoscopic sinus surgery for disease control, bacterial biofilm formation is particularly detrimental, as it reduces the efficacy of surgical intervention. Effective treatments that prevent biofilm formation in post-operative patients in CRS are currently limited. To address this unmet need, we report the controlled release of silver nanoparticles (AgNps) with silk-elastinlike protein-based polymers (SELPs) to prevent bacterial biofilm formation in CRS. This polymeric network is liquid at room temperature and forms a hydrogel at body temperature, and is hence, capable of conforming to the sinonasal cavity upon administration. SELP hydrogels demonstrated sustained AgNp and silver ion release for the studied period of three days, potent in vitro antibacterial activity against Pseudomonas aeruginosa (**** p < 0.0001) and Staphylococcus aureus (**** p < 0.0001), two of the most commonly virulent bacterial strains observed in patients with post-operative CRS, and high cytocompatibility with human nasal epithelial cells. Antibacterial controlled release platform shows promise for treating patients suffering from prolonged sinonasal cavity infections due to biofilms.
Collapse
Affiliation(s)
- Bhuvanesh Yathavan
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; (B.Y.); (T.C.); (A.P.); (J.A.A.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA;
| | - Tanya Chhibber
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; (B.Y.); (T.C.); (A.P.); (J.A.A.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA;
| | - Douglas Steinhauff
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Abigail Pulsipher
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; (B.Y.); (T.C.); (A.P.); (J.A.A.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Otolaryngology—Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Jeremiah A. Alt
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; (B.Y.); (T.C.); (A.P.); (J.A.A.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Otolaryngology—Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Hamidreza Ghandehari
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; (B.Y.); (T.C.); (A.P.); (J.A.A.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Otolaryngology—Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Paris Jafari
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; (B.Y.); (T.C.); (A.P.); (J.A.A.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA;
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
4
|
Liu X, Zhao L, Wu B, Chen F. Improving solubility of poorly water-soluble drugs by protein-based strategy: A review. Int J Pharm 2023; 634:122704. [PMID: 36758883 DOI: 10.1016/j.ijpharm.2023.122704] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/30/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Poorly water-soluble drugs are frequently encountered and present a most challengeable difficulty in pharmaceutical development. Poor solubility of drugs can lead to suboptimal bioavailability and therapeutic efficiency. Increasing efforts have been contributed to improve the solubility of poorly water-soluble drugs for better pharmacokinetics and pharmacodynamics. Among various solubility enhancement technologies, protein-based strategy to address poorly water-soluble drugs issues has special interests for natural advantages including versatile interactions between proteins and hydrophobic drugs, biocompatibility, biodegradation, and metabolization of proteins. The protein-drug formulations could be formed by covalent conjugations or noncovalent interactions to facilitate solubility of poorly water-soluble drugs. This review is to summarize the advances using proteins including plant proteins, mammalian proteins, and recombinant proteins, to enhance water solubility of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Xiaowen Liu
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, China; Shanghai Engineering Research Center of Industrial Asymmetric Catalysis of Chiral Drugs, 220 Handan Road, Shanghai 200433, China.
| | - Limin Zhao
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, China; Shanghai Engineering Research Center of Industrial Asymmetric Catalysis of Chiral Drugs, 220 Handan Road, Shanghai 200433, China
| | - Baojian Wu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Fener Chen
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, 220 Handan Road, Shanghai 200433, China; Shanghai Engineering Research Center of Industrial Asymmetric Catalysis of Chiral Drugs, 220 Handan Road, Shanghai 200433, China.
| |
Collapse
|
5
|
Yuan G, Liu Z, Wang W, Liu M, Xu Y, Hu W, Fan Y, Zhang X, Liu Y, Si G. Multifunctional nanoplatforms application in the transcatheter chemoembolization against hepatocellular carcinoma. J Nanobiotechnology 2023; 21:68. [PMID: 36849981 PMCID: PMC9969656 DOI: 10.1186/s12951-023-01820-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has the sixth-highest new incidence and fourth-highest mortality worldwide. Transarterial chemoembolization (TACE) is one of the primary treatment strategies for unresectable HCC. However, the therapeutic effect is still unsatisfactory due to the insufficient distribution of antineoplastic drugs in tumor tissues and the worsened post-embolization tumor microenvironment (TME, e.g., hypoxia and reduced pH). Recently, using nanomaterials as a drug delivery platform for TACE therapy of HCC has been a research hotspot. With the development of nanotechnology, multifunctional nanoplatforms have been developed to embolize the tumor vasculature, creating conditions for improving the distribution and bioavailability of drugs in tumor tissues. Currently, the researchers are focusing on functionalizing nanomaterials to achieve high drug loading efficacy, thorough vascular embolization, tumor targeting, controlled sustained release of drugs, and real-time imaging in the TACE process to facilitate precise embolization and enable therapeutic procedures follow-up imaging of tumor lesions. Herein, we summarized the recent advances and applications of functionalized nanomaterials based on TACE against HCC, believing that developing these functionalized nanoplatforms may be a promising approach for improving the TACE therapeutic effect of HCC.
Collapse
Affiliation(s)
- Gang Yuan
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Zhiyin Liu
- grid.488387.8Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Weiming Wang
- grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China ,grid.488387.8Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Mengnan Liu
- grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China ,grid.488387.8National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yanneng Xu
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Wei Hu
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Yao Fan
- grid.410578.f0000 0001 1114 4286Department of Anus and Intestine Surgery, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China
| | - Xun Zhang
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China
| | - Yong Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Guangyan Si
- Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
6
|
Garcia Garcia C, Patkar SS, Wang B, Abouomar R, Kiick KL. Recombinant protein-based injectable materials for biomedical applications. Adv Drug Deliv Rev 2023; 193:114673. [PMID: 36574920 DOI: 10.1016/j.addr.2022.114673] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/09/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Injectable nanocarriers and hydrogels have found widespread use in a variety of biomedical applications such as local and sustained biotherapeutic cargo delivery, and as cell-instructive matrices for tissue engineering. Recent advances in the development and application of recombinant protein-based materials as injectable platforms under physiological conditions have made them useful platforms for the development of nanoparticles and tissue engineering matrices, which are reviewed in this work. Protein-engineered biomaterials are highly customizable, and they provide distinctly tunable rheological properties, encapsulation efficiencies, and delivery profiles. In particular, the key advantages of emerging technologies which harness the stimuli-responsive properties of recombinant polypeptide-based materials are highlighted in this review.
Collapse
Affiliation(s)
- Cristobal Garcia Garcia
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Sai S Patkar
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Bin Wang
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Ramadan Abouomar
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE 19176, USA.
| |
Collapse
|
7
|
Stewart RJ, Sima M, Karz J, Jones JP. Material characterization of GPX ®: A versatile in situ solidifying embolic platform technology. Front Bioeng Biotechnol 2023; 11:1095148. [PMID: 36726745 PMCID: PMC9885798 DOI: 10.3389/fbioe.2023.1095148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
Endovascular embolization is a minimally invasive procedure during which blood flow to targeted tissues is selectively occluded. The list of clinical indications for embolization continues to expand. Liquid embolic agents are injectable compositions that transition into a solid or semi-solid form when introduced into blood vessels. The mechanism that triggers the liquid-to-solid transition is a key distinguishing feature of liquid embolic agents. GPX is a waterborne liquid embolic agent comprising oppositely charged polyelectrolytes: polyguanidinum and inorganic polyphoshate. In situ solidification is driven by electrostatic condensation of the polyelectrolytes, triggered by ionic strength differentials. We report in vitro characterization of the material properties of GPX, it is in vivo effectiveness in acute animal studies, and its potential for chemoembolization. The viscosity of GPX can be varied over a wide range by adjusting the polyguanidinium MW and/or concentration. Formulation of GPX with either tantalum microparticles (30 wt%) or iodinated radiocontrast agents (300 mgI ml-1) did not significantly change the flow behavior of GPX; the viscosity was independent of shear rate and remained within a clinically practical range (80-160 cP). Formulation of GPX with doxorubicin substantially increased viscosity at low shear rates and resulted in a power law dependence on shear rate. High contrast and effective vascular occlusion were demonstrated in both swine kidneys and rete mirabile. Contrast from iodinated compounds was temporary, dissipating within hours. The doxorubicin in vitro release profile was linear over 90 days. The results demonstrate that GPX is a versatile liquid embolic platform that can be formulated with a wide range of viscosities injectable at clinically practical flow rates, with either transient or permanent contrast, and that can provide prolonged zero-order delivery of doxorubicin to embolized tissues.
Collapse
Affiliation(s)
- Russell J. Stewart
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States,*Correspondence: Russell J. Stewart,
| | - Monika Sima
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Jessica Karz
- Fluidx Medical Technology, Inc., Salt Lake City, UT, United States
| | - Joshua P. Jones
- Fluidx Medical Technology, Inc., Salt Lake City, UT, United States
| |
Collapse
|
8
|
Griswold E, Cappello J, Ghandehari H. Silk-elastinlike protein-based hydrogels for drug delivery and embolization. Adv Drug Deliv Rev 2022; 191:114579. [PMID: 36306893 DOI: 10.1016/j.addr.2022.114579] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023]
Abstract
Silk-Elastinlike Protein-Based Polymers (SELPs) can form thermoresponsive hydrogels that allow for the generation of in-situ drug delivery matrices. They are produced by recombinant techniques, enabling exact control of monomer sequence and polymer length. In aqueous solutions SELP strands form physical crosslinks as a function of temperature increase without the addition of crosslinking agents. Gelation kinetics, modulus of elasticity, pore size, drug release, biorecognition, and biodegradation of SELP hydrogels can be controlled by placement of amino acid residues at strategic locations in the polymer backbone. SELP hydrogels have been investigated for delivery of a variety of bioactive agents including small molecular weight drugs and fluorescent probes, oligomers of glycosaminoglycans, polymeric macromolecules, proteins, plasmid DNA, and viral gene delivery systems. In this review we provide a background for use of SELPs in matrix-mediated delivery and summarize recent investigations of SELP hydrogels for controlled delivery of bioactive agents as well as their use as liquid embolics.
Collapse
Affiliation(s)
- Ethan Griswold
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center of Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Joseph Cappello
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Hamidreza Ghandehari
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center of Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
9
|
Raut H, Jadhav C, Shetty K, Laxane N, Nijhawan HP, Rao GSNK, Alavala RR, Joshi G, Patro CN, Soni G, Yadav KS. Sorafenib tosylate novel drug delivery systems: implications of nanotechnology in both approved and unapproved indications. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
10
|
Breuer JA, Ahmed KH, Al-Khouja F, Macherla AR, Muthoka JM, Abi-Jaoudeh N. Interventional oncology: new techniques and new devices. Br J Radiol 2022; 95:20211360. [PMID: 35731848 PMCID: PMC9815742 DOI: 10.1259/bjr.20211360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 01/13/2023] Open
Abstract
Interventional oncology is a rapidly emerging field in the treatment of cancer. Minimally invasive techniques such as transarterial embolization with chemotherapeutic and radioactive agents are established therapies and are found in multiple guidelines for the management of primary and metastatic liver lesions. Percutaneous ablation is also an alternative to surgery for small liver, renal, and pancreatic tumors. Recent research in the niche of interventional oncology has focused on improving outcomes of established techniques in addition to the development of novel therapies. In this review, we address the recent and current advancements in devices, technologies, and techniques of chemoembolization and ablation: thermal ablation, histotripsy, high-intensity focused ultrasound, embolization strategies, liquid embolic agents, and local immunotherapy/antiviral therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Nadine Abi-Jaoudeh
- Department of Radiological Sciences, University of California Irvine, Orange, USA
| |
Collapse
|
11
|
Ko G, Choi JW, Lee N, Kim D, Hyeon T, Kim HC. Recent progress in liquid embolic agents. Biomaterials 2022; 287:121634. [PMID: 35716628 DOI: 10.1016/j.biomaterials.2022.121634] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/21/2022]
Abstract
Vascular embolization is a non-surgical procedure used to treat diseases or morbid conditions related to blood vessels, such as bleeding, arteriovenous malformation, aneurysm, and hypervascular tumors, through the intentional occlusion of blood vessels. Among various types of embolic agents that have been applied, liquid embolic agents are gaining an increasing amount of attention owing to their advantages in distal infiltration into regions where solid embolic agents cannot reach, enabling more extensive embolization. Meanwhile, recent advances in biomaterials and technologies have also contributed to the development of novel liquid embolic agents that can resolve the challenges faced while using the existing embolic materials. In this review, we briefly summarize the clinically used embolic agents and their applications, and then present selected research results that overcome the limitations of the embolic agents in use. Through this review, we suggest the required properties of liquid embolic agents that ensure efficacy, which can replace the existing agents, providing directions for the future development in this field.
Collapse
Affiliation(s)
- Giho Ko
- Center for Nanoparticle Research, Institute for Basic Spegcience (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin Woo Choi
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Nohyun Lee
- School of Advanced Materials Engineering, Kookmin University, Seoul 02707, Republic of Korea
| | - Dokyoon Kim
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan 15588, Republic of Korea.
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Spegcience (IBS), Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea.
| | - Hyo-Cheol Kim
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
12
|
Jia G, Van Valkenburgh J, Chen AZ, Chen Q, Li J, Zuo C, Chen K. Recent advances and applications of microspheres and nanoparticles in transarterial chemoembolization for hepatocellular carcinoma. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1749. [PMID: 34405552 PMCID: PMC8850537 DOI: 10.1002/wnan.1749] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022]
Abstract
Transarterial chemoembolization (TACE) is a recommended treatment for patients suffering from intermediate and advanced hepatocellular carcinoma (HCC). As compared to the conventional TACE, drug-eluting bead TACE demonstrates several advantages in terms of survival, treatment response, and adverse effects. The selection of embolic agents is critical to the success of TACE. Many studies have been performed on the modification of the structure, size, homogeneity, biocompatibility, and biodegradability of embolic agents. Continuing efforts are focused on efficient loading of versatile chemotherapeutics, controlled sizes for sufficient occlusion, real-time detection intra- and post-procedure, and multimodality imaging-guided precise treatment. Here, we summarize recent advances and applications of microspheres and nanoparticles in TACE for HCC. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Guorong Jia
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Nuclear Medicine, Changhai Hospital of Shanghai, Shanghai, China
| | - Juno Van Valkenburgh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Austin Z. Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Quan Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jindian Li
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Changjing Zuo
- Department of Nuclear Medicine, Changhai Hospital of Shanghai, Shanghai, China
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
13
|
Hatlevik Ø, Jensen M, Steinhauff D, Wei X, Huo E, Jedrzkiewicz J, Cappello J, Cheney D, Ghandehari H. Translational Development of a Silk-Elastinlike Protein Polymer Embolic for Transcatheter Arterial Embolization. Macromol Biosci 2022; 22:e2100401. [PMID: 34978152 PMCID: PMC9007042 DOI: 10.1002/mabi.202100401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/08/2021] [Indexed: 02/03/2023]
Abstract
Locally blocking blood flow to tumors with embolic materials is the key to transcatheter arterial embolization for treating hepatocellular carcinoma. Current microparticle agents do not deeply penetrate target tissues and are compatible with a very limited selection of therapeutic agents. Silk-elastinlike protein polymers (SELPs) combine the solubility of elastin and the strength of silk to create an easily injected liquid embolic that transition into a solid depot amenable to loading with drugs, gene therapy agents, or biologics. SELP, injected as liquid solution, penetrates the vasculature before transitioning to a solid hydrogel. The objective of this manuscript is to evaluate SELP embolization, stability, and biocompatibility at 7-, 30-, and 90-day survival intervals in a porcine model. SELP embolics selectively block blood flow in the kidneys and livers, with no off-target infarctions. As assessed with angiography, SELP renal embolization exhibits decreasing persistence for the duration of the 90-day study period. There is an increased presence of microscopic SELP emboli in the renal setting, compared to Embosphere. Histologically scored inflammatory reactions to SELP are decreased in both the renal and hepatic implantations compared to Embosphere. In conclusion, a bioresorbable SELP liquid embolic system deeply penetrates target tissue and selectively embolizes blood vessels in vivo.
Collapse
Affiliation(s)
| | | | | | - Xiaomei Wei
- TheraTarget Inc. 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA
| | - Eugene Huo
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 884112, USA, Department of Radiology & Biomedical Imaging, University of California San Francisco, 505 Parnassus, San Francisco, CA 94143, USA
| | - Jolanta Jedrzkiewicz
- Department of Pathology and ARUP Laboratories, University of Utah, School of Medicine, 30 N 1900 E, Salt Lake City, UT 84132, USA
| | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 884112, USA
| | - Darwin Cheney
- TheraTarget Inc. 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA, Utah Center for Nanomedicine, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT 84112, USA
| | | |
Collapse
|
14
|
Steinhauff D, Jensen MM, Griswold E, Jedrzkiewicz J, Cappello J, Oottamasathien S, Ghandehari H. An Oligomeric Sulfated Hyaluronan and Silk-Elastinlike Polymer Combination Protects against Murine Radiation Induced Proctitis. Pharmaceutics 2022; 14:pharmaceutics14010175. [PMID: 35057068 PMCID: PMC8777937 DOI: 10.3390/pharmaceutics14010175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 01/23/2023] Open
Abstract
Semisynthetic glycosaminoglycan ethers (SAGEs) are short, sulfated hyaluronans which combine the natural properties of hyaluronan with chemical sulfation. In a murine model, SAGEs provide protection against radiation induced proctitis (RIP), a side effect of lower abdominal radiotherapy for cancer. The anti-inflammatory effects of SAGE have been studied in inflammatory diseases at mucosal barrier sites; however, few mechanisms have been uncovered necessitating high throughput methods. SAGEs were combined with silk-elastinlike polymers (SELPs) to enhance rectal accumulation in mice. After high radiation exposure to the lower abdominal area, mice were followed for 3 days or until they met humane endpoints, before evaluation of behavioral pain responses and histological assessment of rectal inflammation. RNA sequencing was conducted on tissues from the 3-day cohort to determine molecular mechanisms of SAGE–SELP. After 3 days, mice receiving the SAGE–SELP combination yielded significantly lowered pain responses and amelioration of radiation-induced rectal inflammation. Mice receiving the drug–polymer combination survived 60% longer than other irradiated mice, with a fraction exhibiting long term survival. Sequencing reveals varied regulation of toll like receptors, antioxidant activities, T-cell signaling, and pathways associated with pain. This investigation elucidates several molecular mechanisms of SAGEs and exhibits promising measures for prevention of RIP.
Collapse
Affiliation(s)
- Douglas Steinhauff
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; (D.S.); (E.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Mark Martin Jensen
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (M.M.J.); (S.O.)
| | - Ethan Griswold
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; (D.S.); (E.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA;
| | - Siam Oottamasathien
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (M.M.J.); (S.O.)
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Hamidreza Ghandehari
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; (D.S.); (E.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA;
- Correspondence:
| |
Collapse
|
15
|
Liu M, Wu C, Ke L, Li Z, Wu YL. Emerging Biomaterials-Based Strategies for Inhibiting Vasculature Function in Cancer Therapy. SMALL METHODS 2021; 5:e2100347. [PMID: 34927997 DOI: 10.1002/smtd.202100347] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Indexed: 06/14/2023]
Abstract
The constant feeding of oxygen and nutrients through the blood vasculature has a vital role in maintaining tumor growth. Interestingly, recent endeavors have shown that nanotherapeutics with the strategy to block tumor blood vessels feeding nutrients and oxygen for starvation therapy can be helpful in cancer treatment. However, this field has not been detailed. Hence, this review will present an exhaustive summary of the existing biomaterial based strategies to disrupt tumor vascular function for effective cancer treatment, including hydrogel or nanogel-mediated local arterial embolism, thrombosis activator loaded nano-material-mediated vascular occlusion and anti-vascular drugs that block tumor vascular function, which may be beneficial to the design of anti-cancer nanomedicine by targeting the tumor vascular system.
Collapse
Affiliation(s)
- Minting Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Lingjie Ke
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhiguo Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
16
|
Steinhauff D, Jensen M, Talbot M, Jia W, Isaacson K, Jedrzkiewicz J, Cappello J, Oottamasathien S, Ghandehari H. Silk-elastinlike copolymers enhance bioaccumulation of semisynthetic glycosaminoglycan ethers for prevention of radiation induced proctitis. J Control Release 2021; 332:503-515. [PMID: 33691185 DOI: 10.1016/j.jconrel.2021.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/17/2021] [Accepted: 03/01/2021] [Indexed: 12/22/2022]
Abstract
Radiation-induced proctitis (RIP) is a debilitating adverse event that occurs commonly during lower abdominal radiotherapy. The lack of prophylactic treatment strategies leads to diminished patient quality of life, disruption of radiotherapy schedules, and limitation of radiotherapy efficacy due to dose-limiting toxicities. Semisynthetic glycosaminoglycan ethers (SAGE) demonstrate protective effects from RIP. However, low residence time in the rectal tissue limits their utility. We investigated controlled delivery of GM-0111, a SAGE analogue with demonstrated efficacy against RIP, using a series of temperature-responsive polymers to compare how distinct phase change behaviors, mechanical properties and release kinetics influence rectal bioaccumulation. Poly(lactic acid)-co-(glycolic acid)-block-poly(ethylene glycol)-block-poly(lactic acid)-co-(glycolic acid) copolymers underwent macroscopic phase separation, expelling >50% of drug during gelation. Poloxamer compositions released GM-0111 cargo within 1 h, while silk-elastinlike copolymers (SELPs) enabled controlled release over a period of 12 h. Bioaccumulation was evaluated using fluorescence imaging and confocal microscopy. SELP-415K, a SELP analogue with 4 silk units, 15 elastin units, and one elastin unit with lysine residues in the monomer repeats, resulted in the highest rectal bioaccumulation. SELP-415K GM-0111 compositions were then used to provide localized protection from radiation induced tissue damage in a murine model of RIP. Rectal delivery of SAGE using SELP-415K significantly reduced behavioral pain responses, and reduced animal mass loss compared to irradiated controls or treatment with traditional delivery approaches. Histological scoring showed RIP injury was ameliorated for animals treated with GM-0111 delivered by SELP-415K. The enhanced bioaccumulation provided by thermoresponsive SELPs via a liquid to semisolid transition improved rectal delivery of GM-0111 to mice and radioprotection in a RIP model.
Collapse
Affiliation(s)
- D Steinhauff
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - M Jensen
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - M Talbot
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - W Jia
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - K Isaacson
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - J Jedrzkiewicz
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - J Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - S Oottamasathien
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - H Ghandehari
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
17
|
Ahiwale RJ, Chellampillai B, Pawar AP. Investigation of novel sorafenib tosylate loaded biomaterial based nano-cochleates dispersion system for treatment of hepatocellular carcinoma. J DISPER SCI TECHNOL 2021. [DOI: 10.1080/01932691.2021.1878034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Raj J. Ahiwale
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth University, Pune, Maharashtra, India
| | - Bothiraja Chellampillai
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth University, Pune, Maharashtra, India
| | - Atmaram P. Pawar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth University, Pune, Maharashtra, India
| |
Collapse
|
18
|
Transarterial Embolization of Liver Cancer in a Transgenic Pig Model. J Vasc Interv Radiol 2021; 32:510-517.e3. [PMID: 33500185 DOI: 10.1016/j.jvir.2020.09.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/27/2022] Open
Abstract
PURPOSE To develop and characterize a porcine model of liver cancer that could be used to test new locoregional therapies. MATERIALS AND METHODS Liver tumors were induced in 18 Oncopigs (transgenic pigs with Cre-inducible TP53R167H and KRASG12D mutations) by using an adenoviral vector encoding the Cre-recombinase gene. The resulting 60 tumors were characterized on multiphase contrast-enhanced CT, angiography, perfusion, micro-CT, and necropsy. Transarterial embolization was performed using 40-120 μm (4 pigs) or 100-300 μm (4 pigs) Embosphere microspheres. Response to embolization was evaluated on imaging. Complications were determined based on daily clinical evaluation, laboratory results, imaging, and necropsy. RESULTS Liver tumors developed at 60/70 (86%) inoculated sites. Mean tumor size was 2.1 cm (range, 0.3-4 cm) at 1 week. Microscopically, all animals developed poorly differentiated to undifferentiated carcinomas accompanied by a major inflammatory component, which resembled undifferentiated carcinomas of the human pancreatobiliary tract. Cytokeratin and vimentin expression confirmed epithelioid and mesenchymal differentiation, respectively. Lymph node, lung, and peritoneal metastases were seen in some cases. On multiphase CT, all tumors had a hypovascular center, and 17/60 (28%) had a hypervascular rim. After transarterial embolization, noncontrast CT showed retained contrast medium in the tumors. Follow-up contrast-enhanced scan showed reduced size of tumors after embolization using either 40-120 μm or 100-300 μm Embosphere microspheres, while untreated tumors showed continued growth. CONCLUSIONS Liver tumors can be induced in a transgenic pig and can be successfully treated using bland embolization.
Collapse
|
19
|
Chambre L, Martín-Moldes Z, Parker RN, Kaplan DL. Bioengineered elastin- and silk-biomaterials for drug and gene delivery. Adv Drug Deliv Rev 2020; 160:186-198. [PMID: 33080258 PMCID: PMC7736173 DOI: 10.1016/j.addr.2020.10.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/30/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022]
Abstract
Advances in medical science have led to diverse new therapeutic modalities, as well as enhanced understanding of the progression of various disease states. These findings facilitate the design and development of more customized and exquisite drug delivery systems that aim to improve therapeutic indices of drugs to treat a variety of conditions. Synthetic polymer-based drug carriers have often been the focus of such research. However, these structures suffer from challenges with heterogeneity of the starting material, limited chemical features, complex functionalization methods, and in some cases a lack of biocompatibility. Consequently, protein-based polymers have garnered much attention in recent years due to their monodisperse features, ease of production and functionalization, and biocompatibility. Genetic engineering techniques enable the advancement of protein-based drug delivery systems with finely tuned physicochemical properties, and thus an expanded level of customization unavailable with synthetic polymers. Of these genetically engineered proteins, elastin-like proteins (ELP), silk-like proteins (SLP), and silk-elastin-like proteins (SELP) provide a unique set of alternatives for designing drug delivery systems due to their inherent chemical and physical properties and ease of engineering afforded by recombinant DNA technologies. In this review we examine the advantages of genetically engineered drug delivery systems with emphasis on ELP and SLP constructions. Methods for fabrication and relevant biomedical applications will also be discussed.
Collapse
Affiliation(s)
- Laura Chambre
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Zaira Martín-Moldes
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Rachael N Parker
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA.
| |
Collapse
|
20
|
Isaacson KJ, Jensen MM, Steinhauff DB, Kirklow JE, Mohammadpour R, Grunberger JW, Cappello J, Ghandehari H. Location of stimuli-responsive peptide sequences within silk-elastinlike protein-based polymers affects nanostructure assembly and drug-polymer interactions. J Drug Target 2020; 28:766-779. [PMID: 32306773 DOI: 10.1080/1061186x.2020.1757099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Silk-elastinlike protein polymers (SELPs) self-assemble into nanostructures when designed with appropriate silk-to-elastin ratios. Here, we investigate the effect of insertion of a matrix metalloproteinase-responsive peptide sequence, GPQGIFGQ, into various locations within the SELP backbone on supramolecular self-assembly. Insertion of the hydrophilic, enzyme-degradable sequence into the elastin repeats allows the formation of dilution-stable nanostructures, while insertion into the hydrophobic silk motifs inhibited self-assembly. The SELP assemblies retained their lower critical solution temperature (LCST) thermal response, allowing up to eightfold volumetric changes due to temperature-induced size change. A model hydrophobic drug was incorporated into SELP nanoassemblies utilising a combination of precipitation, incubation and tangential flow filtration. While the nanoconstructs degraded in response to MMP activity, drug release kinetics was independent of MMP concentration. Drug release modelling suggests that release is driven by rates of water penetration into the SELP nanostructures and drug dissolution. In vitro testing revealed that SELP nanoassemblies reduced the immunotoxic and haemolytic side effects of doxorubicin in human blood while maintaining its cytotoxic activity.
Collapse
Affiliation(s)
- Kyle J Isaacson
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - M Martin Jensen
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Douglas B Steinhauff
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - James E Kirklow
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Raziye Mohammadpour
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Jason W Grunberger
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
21
|
Tahir N, Madni A, Li W, Correia A, Khan MM, Rahim MA, Santos HA. Microfluidic fabrication and characterization of Sorafenib-loaded lipid-polymer hybrid nanoparticles for controlled drug delivery. Int J Pharm 2020; 581:119275. [PMID: 32229283 DOI: 10.1016/j.ijpharm.2020.119275] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 01/02/2023]
Abstract
Lipid polymer hybrid nanoparticles (LPHNPs) have been merged as potential nanocarriers for treatment of cancer. In the present study, LPHNPs loaded with Sorafenib (SFN) were prepared with PLGA, Lecithin and DSPE-PEG 2000 by using the bulk nanoprecipitation and microfluidic (MF) co-flow nanoprecipitation techniques. Herein, a glass capillary microfluidic device was primed to optimize the LPHNPs and compared to the bulk nanoprecipitation method. The morphological analysis of prepared LPHNPs revealed the well-defined spherical nano-sized particles in bulk nanoprecipitation method. Whereas, core shell morphology was observed in the MF method. The formulation prepared by the MF method (MF1-MF3) indicated relatively higher % EE (95.0%, 93.8% and 88.7%) and controlled release of the SFN from the particles as compared to the LPHNPs obtained by the bulk nanoprecipitation method. However, the release of SFN from all LPHNP formulation followed Higuchi model (R2 = 0.9901-0.9389) with Fickian diffusion mechanism. Fourier transform infrared spectroscopy (FTIR), Differential scanning calorimetry (DSC) and powder X-rays diffraction (pXRD) studies depicted the compatibility of SFN with all the structural components. In addition, the colloidal stability, in vitro cytotoxicity and cell growth inhibition studies of LPHNPs also demonstrated stability in biological media, biocompatibility and safety with enhanced anti-proliferative effects than the free SFN in breast cancer and prostate cancer cells. In conclusion, LPHNPs provided a prospective platform for the cancer chemotherapy and substantially improved the knowledge of fabrication and optimization of the hybrid nanoparticles.
Collapse
Affiliation(s)
- Nayab Tahir
- College of Pharmacy, University of Sargodha, Sargodha, Pakistan; Department of Pharmacy, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Asadullah Madni
- Department of Pharmacy, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan.
| | - Wei Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Muhammad Muzamil Khan
- Department of Pharmacy, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan
| | - Muhammad Abdur Rahim
- Department of Pharmacy, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|
22
|
Jensen MM, Barber ZB, Khurana N, Isaacson KJ, Steinhauff D, Green B, Cappello J, Pulsipher A, Ghandehari H, Alt JA. A dual-functional Embolization-Visualization System for Fluorescence image-guided Tumor Resection. Theranostics 2020; 10:4530-4543. [PMID: 32292513 PMCID: PMC7150499 DOI: 10.7150/thno.39700] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Intraoperative bleeding impairs physicians' ability to visualize the surgical field, leading to increased risk of surgical complications and reduced outcomes. Bleeding is particularly challenging during endoscopic-assisted surgical resection of hypervascular tumors in the head and neck. A tool that controls bleeding while marking tumor margins has the potential to improve gross tumor resection, reduce surgical morbidity, decrease blood loss, shorten procedure time, prevent damage to surrounding tissues, and limit postoperative pain. Herein, we develop and characterize a new system that combines pre-surgical embolization with improved visualization for endoscopic fluorescence image-guided tumor resection. Methods: Silk-elastinlike protein (SELP) polymers were employed as liquid embolic vehicles for delivery of a clinically used near-infrared dye, indocyanine green (ICG). The biophysical properties of SELP, including gelation kinetics, modulus of elasticity, and viscosity, in response to ICG incorporation using rheology, were characterized. ICG release from embolic SELP was modeled in tissue phantoms and via fluorescence imaging. The embolic capability of the SELP-ICG system was then tested in a microfluidic model of tumor vasculature. Lastly, the cytotoxicity of the SELP-ICG system in L-929 fibroblasts and human umbilical vein endothelial cells (HUVEC) was assessed. Results: ICG incorporation into SELP accelerated gelation and increased its modulus of elasticity. The SELP embolic system released 83 ± 8% of the total ICG within 24 hours, matching clinical practice for pre-surgical embolization procedures. Adding ICG to SELP did not reduce injectability, but did improve the gelation kinetics. After simulated embolization, ICG released from SELP in tissue phantoms diffused a sufficient distance to deliver dye throughout a tumor. ICG-loaded SELP was injectable through a clinical 2.3 Fr microcatheter and demonstrated deep penetration into 50-µm microfluidic-simulated blood vessels with durable occlusion. Incorporation of ICG into SELP improved biocompatibility with HUVECs, but had no effect on L-929 cell viability. Principle Conclusions: We report the development and characterization of a new, dual-functional embolization-visualization system for improving fluorescence-imaged endoscopic surgical resection of hypervascular tumors.
Collapse
Affiliation(s)
- M. Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Zachary B. Barber
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Nitish Khurana
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112 USA
| | - Kyle J. Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Douglas Steinhauff
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Bryant Green
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112 USA
| | - Abigail Pulsipher
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84113
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84113
| | - Jeremiah A. Alt
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84113
| |
Collapse
|
23
|
Sun Z, Song C, Wang C, Hu Y, Wu J. Hydrogel-Based Controlled Drug Delivery for Cancer Treatment: A Review. Mol Pharm 2020; 17:373-391. [PMID: 31877054 DOI: 10.1021/acs.molpharmaceut.9b01020] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
As an emerging drug carrier, hydrogels have been widely used for tumor drug delivery. A hydrogel drug carrier can cause less severe side effects than systemic chemotherapy and can achieve sustained delivery of a drug at tumor sites. In addition, hydrogels have excellent biocompatibility and biodegradability and lower toxicity than nanoparticle carriers. Smart hydrogels can respond to stimuli in the environment (e.g., heat, pH, light, and ultrasound), enabling in situ gelation and controlled drug release, which greatly enhance the convenience and efficiency of drug delivery. Here, we summarize the different sizes of hydrogels used for cancer treatment and their related delivery routes, discuss the design strategies for stimuli-responsive hydrogels, and review the research concerning smart hydrogels reported in the past few years.
Collapse
Affiliation(s)
- Zhaoyi Sun
- School of Chemistry and Chemical Engineering , Nanjing University , 210046 Nanjing , China
| | - Chengjun Song
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences , Nanjing University , 210093 Nanjing , China
| | - Chao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences , Nanjing University , 210093 Nanjing , China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences , Nanjing University , 210093 Nanjing , China.,Jiangsu Key Laboratory for Nano Technology , Nanjing University , 210093 Nanjing , China.,Institute of Drug R&D , Medical School of Nanjing University , 210093 Nanjing , China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences , Nanjing University , 210093 Nanjing , China.,Jiangsu Key Laboratory for Nano Technology , Nanjing University , 210093 Nanjing , China.,Institute of Drug R&D , Medical School of Nanjing University , 210093 Nanjing , China
| |
Collapse
|
24
|
Cirillo G, Spizzirri UG, Curcio M, Nicoletta FP, Iemma F. Injectable Hydrogels for Cancer Therapy over the Last Decade. Pharmaceutics 2019; 11:E486. [PMID: 31546921 PMCID: PMC6781516 DOI: 10.3390/pharmaceutics11090486] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/10/2019] [Accepted: 09/17/2019] [Indexed: 01/07/2023] Open
Abstract
The interest in injectable hydrogels for cancer treatment has been significantly growing over the last decade, due to the availability of a wide range of starting polymer structures with tailored features and high chemical versatility. Many research groups are working on the development of highly engineered injectable delivery vehicle systems suitable for combined chemo-and radio-therapy, as well as thermal and photo-thermal ablation, with the aim of finding out effective solutions to overcome the current obstacles of conventional therapeutic protocols. Within this work, we have reviewed and discussed the most recent injectable hydrogel systems, focusing on the structure and properties of the starting polymers, which are mainly classified into natural or synthetic sources. Moreover, mapping the research landscape of the fabrication strategies, the main outcome of each system is discussed in light of possible clinical applications.
Collapse
Affiliation(s)
- Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy
| | - Umile Gianfranco Spizzirri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy.
| | - Manuela Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy.
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy.
| | - Francesca Iemma
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy.
| |
Collapse
|
25
|
Wongpinyochit T, Vassileiou AD, Gupta S, Mushrif SH, Johnston BF, Seib FP. Unraveling the Impact of High-Order Silk Structures on Molecular Drug Binding and Release Behaviors. J Phys Chem Lett 2019; 10:4278-4284. [PMID: 31318218 DOI: 10.1021/acs.jpclett.9b01591] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Silk continues to amaze: over the past decade, new research threads have emerged that include the use of silk fibroin for advanced pharmaceutics, including its suitability for drug delivery. Despite this ongoing interest, the details of silk fibroin structures and their subsequent drug interactions at the molecular level remain elusive, primarily because of the difficulties encountered in modeling the silk fibroin molecule. Here, we generated an atomistic silk model containing amorphous and crystalline regions. We then exploited advanced well-tempered metadynamics simulations to generate molecular conformations that we subsequently exposed to classical molecular dynamics simulations to monitor both drug binding and release. Overall, this study demonstrated the importance of the silk fibroin primary sequence, electrostatic interactions, hydrogen bonding, and higher-order conformation in the processes of drug binding and release.
Collapse
Affiliation(s)
- Thidarat Wongpinyochit
- Strathclyde Institute of Pharmacy and Biomedical Sciences , University of Strathclyde , 161 Cathedral Street , Glasgow G4 0RE , United Kingdom
| | - Antony D Vassileiou
- Strathclyde Institute of Pharmacy and Biomedical Sciences , University of Strathclyde , 161 Cathedral Street , Glasgow G4 0RE , United Kingdom
| | - Sukriti Gupta
- Energy Research Institute @ NTU, Interdisciplinary Graduate School , Nanyang Technological University , 50 Nanyang Drive , Singapore 637553
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459
| | - Samir H Mushrif
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459
- Department of Chemical and Materials Engineering , University of Alberta , 9211-116 Street Northwest , Edmonton , Alberta T6G 1H9 , Canada
| | - Blair F Johnston
- Strathclyde Institute of Pharmacy and Biomedical Sciences , University of Strathclyde , 161 Cathedral Street , Glasgow G4 0RE , United Kingdom
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , United Kingdom
| | - F Philipp Seib
- Strathclyde Institute of Pharmacy and Biomedical Sciences , University of Strathclyde , 161 Cathedral Street , Glasgow G4 0RE , United Kingdom
- Leibniz Institute of Polymer Research Dresden , Max Bergmann Center of Biomaterials Dresden , Hohe Strasse 6 , 01069 Dresden , Germany
| |
Collapse
|
26
|
Hu J, Albadawi H, Oklu R, Chong BW, Deipolyi AR, Sheth RA, Khademhosseini A. Advances in Biomaterials and Technologies for Vascular Embolization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1901071. [PMID: 31168915 PMCID: PMC7014563 DOI: 10.1002/adma.201901071] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/24/2019] [Indexed: 05/03/2023]
Abstract
Minimally invasive transcatheter embolization is a common nonsurgical procedure in interventional radiology used for the deliberate occlusion of blood vessels for the treatment of diseased or injured vasculature. A wide variety of embolic agents including metallic coils, calibrated microspheres, and liquids are available for clinical practice. Additionally, advances in biomaterials, such as shape-memory foams, biodegradable polymers, and in situ gelling solutions have led to the development of novel preclinical embolic agents. The aim here is to provide a comprehensive overview of current and emerging technologies in endovascular embolization with respect to devices, materials, mechanisms, and design guidelines. Limitations and challenges in embolic materials are also discussed to promote advancement in the field.
Collapse
Affiliation(s)
- Jingjie Hu
- Division of Vascular & Interventional Radiology, Minimally Invasive Therapeutics Laboratory, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Hassan Albadawi
- Division of Vascular & Interventional Radiology, Minimally Invasive Therapeutics Laboratory, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Rahmi Oklu
- Division of Vascular & Interventional Radiology, Minimally Invasive Therapeutics Laboratory, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Brian W Chong
- Departments of Radiology and Neurological Surgery, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, Arizona 85259, USA
| | - Amy R. Deipolyi
- Department of Interventional Radiology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical Center, 1275 York Avenue, New York, New York 10065, USA
| | - Rahul A. Sheth
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Ali Khademhosseini
- Department of Bioengineering, Department of Radiological Sciences, Department of Chemical and Biomolecular Engineering, Center for Minimally Invasive Therapeutics, California Nanosystems Institute, University of California, 410 Westwood Plaza, Los Angeles, California 90095, USA
| |
Collapse
|
27
|
Jensen MM, Jia W, Schults AJ, Isaacson KJ, Steinhauff D, Green B, Zachary B, Cappello J, Ghandehari H, Oottamasathien S. Temperature-responsive silk-elastinlike protein polymer enhancement of intravesical drug delivery of a therapeutic glycosaminoglycan for treatment of interstitial cystitis/painful bladder syndrome. Biomaterials 2019; 217:119293. [PMID: 31276948 DOI: 10.1016/j.biomaterials.2019.119293] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 12/28/2022]
Abstract
Interstitial cystitis (IC), also known as painful bladder syndrome, is a debilitating chronic condition with many patients failing to respond to current treatment options. Rapid clearance, mucosal coating, and tight epithelium create strong natural barriers that reduce the effectiveness of many pharmacological interventions in the bladder. Intravesical drug delivery (IDD) is the administration of therapeutic compounds or devices to the urinary bladder via a urethral catheter. Previous work in improving IDD for IC has focused on the sustained delivery of analgesics within the bladder and other small molecule drugs which do not address underlying inflammation and bladder damage. Therapeutic glycosaminoglycans (GAG) function by restoring the mucosal barrier within the bladder, promoting healing responses, and preventing irritating solutes from reaching the bladder wall. There is an unmet medical need for a therapy that provides both acute relief of symptoms while alleviating underlying physiological sources of inflammation and promoting healing within the urothelium. Semi-synthetic glycosaminoglycan ethers (SAGE) are an emerging class of therapeutic GAG with intrinsic anti-inflammatory and analgesic properties. To reduce SAGE clearance and enhance its accumulation in the bladder, we developed a silk-elastinlike protein polymer (SELP) based system to enhance SAGE IDD. We evaluated in vitro release kinetics, rheological properties, impact on bladder function, pain response, and bladder inflammation and compared their effectiveness to other temperature-responsive polymers including Poloxamer 407 and poly(lactic-co-glycolic acid)-poly(ethylene glycol). SAGE delivered via SELP-enhanced intravesical delivery substantially improved SAGE accumulation in the urothelium, provided a sustained analgesic effect 24 h after administration, and reduced inflammation.
Collapse
Affiliation(s)
- M Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA; (b)Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112, USA
| | - Wanjian Jia
- Division of Urology, Section of Pediatric Urology, University of Utah, Salt Lake City, UT, 84113, USA
| | - Austin J Schults
- Division of Urology, Section of Pediatric Urology, University of Utah, Salt Lake City, UT, 84113, USA
| | - Kyle J Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA; (b)Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112, USA
| | - Douglas Steinhauff
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA; (b)Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112, USA
| | - Bryant Green
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA; (b)Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112, USA
| | - B Zachary
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA; (b)Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Siam Oottamasathien
- Division of Urology, Section of Pediatric Urology, University of Utah, Salt Lake City, UT, 84113, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112, USA; Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT, 84112, USA; Department of Surgery and Division of Pediatric Urology, Primary Children's Hospital, Salt Lake City, UT, 84113, USA; Department of Pediatric Surgery, Division of Pediatric Urology, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
28
|
Wang Y, Katyal P, Montclare JK. Protein-Engineered Functional Materials. Adv Healthc Mater 2019; 8:e1801374. [PMID: 30938924 PMCID: PMC6703858 DOI: 10.1002/adhm.201801374] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 02/25/2019] [Indexed: 12/13/2022]
Abstract
Proteins are versatile macromolecules that can perform a variety of functions. In the past three decades, they have been commonly used as building blocks to generate a range of biomaterials. Owing to their flexibility, proteins can either be used alone or in combination with other functional molecules. Advances in synthetic and chemical biology have enabled new protein fusions as well as the integration of new functional groups leading to biomaterials with emergent properties. This review discusses protein-engineered materials from the perspectives of domain-based designs as well as physical and chemical approaches for crosslinked materials, with special emphasis on the creation of hydrogels. Engineered proteins that organize or template metal ions, bear noncanonical amino acids (NCAAs), and their potential applications, are also reviewed.
Collapse
Affiliation(s)
- Yao Wang
- Department of Chemical and Biomolecular Engineering, New
York University, Tandon School of Engineering, Brooklyn, NY 11201, United
States
| | - Priya Katyal
- Department of Chemical and Biomolecular Engineering, New
York University, Tandon School of Engineering, Brooklyn, NY 11201, United
States
| | - Jin Kim Montclare
- Department of Chemical and Biomolecular Engineering, New
York University, Tandon School of Engineering, Brooklyn, NY 11201, United
States
- Department of Chemistry, New York University, New York, NY
10003, United States
- Department of Biomaterials, New York University College of
Dentistry, New York, NY 10010, United States
- Department of Radiology, New York University School of
Medicine, New York, New York, 10016, United States
| |
Collapse
|
29
|
Jeganathan S, Budziszewski E, Hernandez C, Wu H, Gilbert D, Tavri S, Exner AA. Tunable Polymer Embolic Implant for Vascular Occlusion. ACS Biomater Sci Eng 2019; 5:1849-1856. [DOI: 10.1021/acsbiomaterials.8b01530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Isaacson KJ, Jensen MM, Watanabe AH, Green BE, Correa MA, Cappello J, Ghandehari H. Self-Assembly of Thermoresponsive Recombinant Silk-Elastinlike Nanogels. Macromol Biosci 2018; 18:10.1002/mabi.201700192. [PMID: 28869362 PMCID: PMC5806626 DOI: 10.1002/mabi.201700192] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/19/2017] [Indexed: 12/28/2022]
Abstract
Recombinant silk-elastinlike protein polymers (SELPs) combine the biocompatibility and thermoresponsiveness of human tropoelastin with the strength of silk. Direct control over structure of these monodisperse polymers allows for precise correlation of structure with function. This work describes the fabrication of the first SELP nanogels and evaluation of their physicochemical properties and thermoresponsiveness. Self-assembly of dilute concentrations of SELPs results in nanogels with enhanced stability over micelles due to physically crosslinked beta-sheet silk segments. The nanogels respond to thermal stimuli via size changes and aggregation. Modifying the ratio and sequence of silk to elastin in the polymer backbone results in alterations in critical gel formation concentration, stability, aggregation, size contraction temperature, and thermal reversibility. The nanogels sequester hydrophobic compounds and show promise in delivery of bioactive agents.
Collapse
Affiliation(s)
- Kyle J Isaacson
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
- Department of Bioengineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
| | - Mark Martin Jensen
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
- Department of Bioengineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
| | - Alexandre H Watanabe
- College of Pharmacy, University of Utah, 30 2000 E., Salt Lake City, UT, 84112, USA
| | - Bryant E Green
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
- Department of Bioengineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
| | - Marcelo A Correa
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
- Department of Bioengineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
| | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S. 2000 E., Salt Lake City, UT, 84112, USA
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
- Department of Bioengineering, University of Utah, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S. 2000 E., Salt Lake City, UT, 84112, USA
| |
Collapse
|
31
|
Jensen MM, Jia W, Isaacson KJ, Schults A, Cappello J, Prestwich GD, Oottamasathien S, Ghandehari H. Silk-elastinlike protein polymers enhance the efficacy of a therapeutic glycosaminoglycan for prophylactic treatment of radiation-induced proctitis. J Control Release 2017; 263:46-56. [PMID: 28232224 DOI: 10.1016/j.jconrel.2017.02.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/15/2017] [Accepted: 02/19/2017] [Indexed: 12/17/2022]
Abstract
Radiation-induced proctitis (RIP) is the most common clinical adverse effect for patients receiving radiotherapy as part of the standard course of treatment for ovarian, prostate, colon, and bladder cancers. RIP limits radiation dosage, interrupts treatment, and lowers patients' quality of life. A prophylactic treatment that protects the gastrointestinal tract from deleterious effects of radiotherapy will significantly improve patient quality of life and may allow for higher and more regular doses of radiation therapy. Semi-synthetic glycosaminoglycan (GAG), generated from the sulfation of hyaluronic acid, are anti-inflammatory but have difficulty achieving therapeutic levels in many tissues. To enhance the delivery of GAG, we created an in situ gelling rectal delivery system using silk-elastinlike protein polymers (SELPs). Using solutions of SELP 815K (which contains 6 repeats of blocks comprised of 8 silk-like units, 15 elastin-like units, and 1 lysine-substituted elastin-like unit) with GAG GM-0111, we created an injectable delivery platform that transitioned in <5min from a liquid at room temperature to a hydrogel at body temperature. The hydrogels released 50% of their payload within 30min and enhanced the accumulation of GAG in the rectum compared to traditional enema-based delivery. Using a murine model of radiation-induced proctitis, the prophylactic delivery of a single dose of GAG from a SELP matrix administered prior to irradiation significantly reduced radiation-induced pain after 3, 7, and 21days by 53±4%, 47±10%, and 12±6%, respectively. Matrix-mediated delivery of GAG by SELP represents an innovative method for more effective treatment of RIP and promises to improve quality of life of cancer patients by allowing higher radiotherapy doses with improved safety.
Collapse
Affiliation(s)
- Mark Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84112, USA
| | - Wanjian Jia
- Division of Urology, Section of Pediatric Urology, University of Utah, Salt Lake City, UT 84113, USA
| | - Kyle J Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84112, USA
| | - Austin Schults
- Division of Urology, Section of Pediatric Urology, University of Utah, Salt Lake City, UT 84113, USA
| | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Glenn D Prestwich
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Siam Oottamasathien
- Division of Urology, Section of Pediatric Urology, University of Utah, Salt Lake City, UT 84113, USA; Department of Surgery and Division of Pediatric Urology, Primary Children's Hospital, Salt Lake City, UT 84113, USA.
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84112, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
32
|
Isaacson KJ, Martin Jensen M, Subrahmanyam NB, Ghandehari H. Matrix-metalloproteinases as targets for controlled delivery in cancer: An analysis of upregulation and expression. J Control Release 2017; 259:62-75. [PMID: 28153760 DOI: 10.1016/j.jconrel.2017.01.034] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/18/2017] [Accepted: 01/26/2017] [Indexed: 02/07/2023]
Abstract
While commonly known for degradation of the extracellular matrix, matrix metalloproteinases (MMPs) exhibit broad potential for use in targeting of bioactive and imaging agents in cancer treatment. MMPs are upregulated at all stages of expression in cancers. A comprehensive analysis of published literature on expression of all MMP subtypes at the genetic, protein, and activity levels in normal and diseased tissues indicate targeting applicability in a variety of cancers. This expression significantly increases at advanced cancer stages, providing an improved opportunity for controlled release in higher-stage patients. Since MMPs are integral at every stage of metastasis, MMP roles in cancer are discussed with a focus on MMP distribution and mobility within cells and tumors for cancer targeting applications. Several strategies for MMP utilization in targeting - such as matrix degradation, MMP cleavage, MMP binding, and MMP-induced environmental changes - are addressed.
Collapse
Affiliation(s)
- Kyle J Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - M Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Nithya B Subrahmanyam
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|