51
|
Saiding Q, Zhang Z, Chen S, Xiao F, Chen Y, Li Y, Zhen X, Khan MM, Chen W, Koo S, Kong N, Tao W. Nano-bio interactions in mRNA nanomedicine: Challenges and opportunities for targeted mRNA delivery. Adv Drug Deliv Rev 2023; 203:115116. [PMID: 37871748 DOI: 10.1016/j.addr.2023.115116] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023]
Abstract
Upon entering the biological milieu, nanomedicines swiftly interact with the surrounding tissue fluid, subsequently being enveloped by a dynamic interplay of biomacromolecules, such as carbohydrates, nucleic acids, and cellular metabolites, but with predominant serum proteins within the biological corona. A notable consequence of the protein corona phenomenon is the unintentional loss of targeting ligands initially designed to direct nanomedicines toward particular cells or organs within the in vivo environment. mRNA nanomedicine displays high demand for specific cell and tissue-targeted delivery to effectively transport mRNA molecules into target cells, where they can exert their therapeutic effects with utmost efficacy. In this review, focusing on the delivery systems and tissue-specific applications, we aim to update the nanomedicine population with the prevailing and still enigmatic paradigm of nano-bio interactions, a formidable hurdle in the pursuit of targeted mRNA delivery. We also elucidate the current impediments faced in mRNA therapeutics and, by contemplating prospective avenues-either to modulate the corona or to adopt an 'ally from adversary' approach-aim to chart a course for advancing mRNA nanomedicine.
Collapse
Affiliation(s)
- Qimanguli Saiding
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Zhongyang Zhang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States; The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Shuying Chen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang 311121, China; Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Yumeng Chen
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Yongjiang Li
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Xueyan Zhen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Muhammad Muzamil Khan
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Wei Chen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Seyoung Koo
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang 311121, China; Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| | - Wei Tao
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
52
|
Sato H, Yamada K, Miyake M, Onoue S. Recent Advancements in the Development of Nanocarriers for Mucosal Drug Delivery Systems to Control Oral Absorption. Pharmaceutics 2023; 15:2708. [PMID: 38140049 PMCID: PMC10747340 DOI: 10.3390/pharmaceutics15122708] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Oral administration of active pharmaceutical ingredients is desirable because it is easy, safe, painless, and can be performed by patients, resulting in good medication adherence. The mucus layer in the gastrointestinal (GI) tract generally acts as a barrier to protect the epithelial membrane from foreign substances; however, in the absorption process after oral administration, it can also disturb effective drug absorption by trapping it in the biological sieve structured by mucin, a major component of mucus, and eliminating it by mucus turnover. Recently, functional nanocarriers (NCs) have attracted much attention due to their immense potential and effectiveness in the field of oral drug delivery. Among them, NCs with mucopenetrating and mucoadhesive properties are promising dosage options for controlling drug absorption from the GI tracts. Mucopenetrating and mucoadhesive NCs can rapidly deliver encapsulated drugs to the absorption site and/or prolong the residence time of NCs close to the absorption membrane, providing better medications than conventional approaches. The surface characteristics of NCs are important factors that determine their functionality, owing to the formation of various kinds of interactions between the particle surface and mucosal components. Thus, a deeper understanding of surface modifications on the biopharmaceutical characteristics of NCs is necessary to develop the appropriate mucosal drug delivery systems (mDDS) for the treatment of target diseases. This review summarizes the basic information and functions of the mucosal layer, highlights the recent progress in designing functional NCs for mDDS, and discusses their performance in the GI tract.
Collapse
Affiliation(s)
- Hideyuki Sato
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (H.S.); (K.Y.)
| | - Kohei Yamada
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (H.S.); (K.Y.)
| | - Masateru Miyake
- Business Integrity and External Affairs, Otsuka Pharmaceutical Co., Ltd., 2-16-4 Konan, Minato-ku, Tokyo 108-8242, Japan;
| | - Satomi Onoue
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (H.S.); (K.Y.)
| |
Collapse
|
53
|
Davis MA, Cho E, Teplensky MH. Harnessing biomaterial architecture to drive anticancer innate immunity. J Mater Chem B 2023; 11:10982-11005. [PMID: 37955201 DOI: 10.1039/d3tb01677c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Immunomodulation is a powerful therapeutic approach that harnesses the body's own immune system and reprograms it to treat diseases, such as cancer. Innate immunity is key in mobilizing the rest of the immune system to respond to disease and is thus an attractive target for immunomodulation. Biomaterials have widely been employed as vehicles to deliver immunomodulatory therapeutic cargo to immune cells and raise robust antitumor immunity. However, it is key to consider the design of biomaterial chemical and physical structure, as it has direct impacts on innate immune activation and antigen presentation to stimulate downstream adaptive immunity. Herein, we highlight the widespread importance of structure-driven biomaterial design for the delivery of immunomodulatory cargo to innate immune cells. The incorporation of precise structural elements can be harnessed to improve delivery kinetics, uptake, and the targeting of biomaterials into innate immune cells, and enhance immune activation against cancer through temporal and spatial processing of cargo to overcome the immunosuppressive tumor microenvironment. Structural design of immunomodulatory biomaterials will profoundly improve the efficacy of current cancer immunotherapies by maximizing the impact of the innate immune system and thus has far-reaching translational potential against other diseases.
Collapse
Affiliation(s)
- Meredith A Davis
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Ezra Cho
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
| | - Michelle H Teplensky
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA.
- Department of Materials Science and Engineering, Boston University, Boston, Massachusetts, 02215, USA
| |
Collapse
|
54
|
Navarro-Marchal SA, Martín-Contreras M, Castro-Santiago D, del Castillo-Santaella T, Graván P, Jódar-Reyes AB, Marchal JA, Peula-García JM. Effect of the Protein Corona Formation on Antibody Functionalized Liquid Lipid Nanocarriers. Int J Mol Sci 2023; 24:16759. [PMID: 38069079 PMCID: PMC10706289 DOI: 10.3390/ijms242316759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
The main aim of this study is to report basic knowledge on how a protein corona (PC) could affect or modify the way in which multifunctionalized nanoparticles interact with cells. With this purpose, we have firstly optimized the development of a target-specific nanocarrier by coupling a specific fluorescent antibody on the surface of functionalized lipid liquid nanocapsules (LLNCs). Thus, an anti-HER2-FITC antibody (αHER2) has been used, HER2 being a surface receptor that is overexpressed in several tumor cells. Subsequently, the in vitro formation of a PC has been developed using fetal bovine serum supplemented with human fibrinogen. Dynamic Light Scattering (DLS), Nanoparticle Tracking Analysis (NTA), Laser Doppler Electrophoresis (LDE), and Gel Chromatography techniques have been used to assure a complete physico-chemical characterization of the nano-complexes with (LLNCs-αHER2-PC) and without (LLNCs-αHER2) the surrounding PC. In addition, cellular assays were performed to study the cellular uptake and the specific cellular-nanocarrier interactions using the SKBR3 (high expression of HER2) breast cancer cell line and human dermal fibroblasts (HDFa) (healthy cell line without expression of HER2 receptors as control), showing that the SKBR3 cell line had a higher transport rate (50-fold) than HDFa at 60 min with LLNCs-αHER2. Moreover, the SKBR3 cell line incubated with LLNCs-αHER2-PC suffered a significant reduction (40%) in the uptake. These results suggest that the formation of a PC onto LLNCs does not prevent specific cell targeting, although it does have an important influence on cell uptake.
Collapse
Affiliation(s)
- Saúl A. Navarro-Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (S.A.N.-M.); (P.G.); (J.A.M.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Excellence Research Unit Modeling Nature (MNat), University of Granada, 18071 Granada, Spain;
| | - Marina Martín-Contreras
- Department of Applied Physics, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - David Castro-Santiago
- Department of Applied Physics, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Teresa del Castillo-Santaella
- Department of Physical Chemistry, Faculty of Pharmacy, University of Granada, 18011 Granada, Spain;
- Biocolloid and Fluid Physics Group, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Pablo Graván
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (S.A.N.-M.); (P.G.); (J.A.M.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Excellence Research Unit Modeling Nature (MNat), University of Granada, 18071 Granada, Spain;
- Biocolloid and Fluid Physics Group, Faculty of Sciences, University of Granada, 18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Ana Belén Jódar-Reyes
- Excellence Research Unit Modeling Nature (MNat), University of Granada, 18071 Granada, Spain;
- Department of Applied Physics, Faculty of Sciences, University of Granada, 18071 Granada, Spain
- Biocolloid and Fluid Physics Group, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; (S.A.N.-M.); (P.G.); (J.A.M.)
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Excellence Research Unit Modeling Nature (MNat), University of Granada, 18071 Granada, Spain;
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - José Manuel Peula-García
- Biocolloid and Fluid Physics Group, Faculty of Sciences, University of Granada, 18071 Granada, Spain
- Department of Applied Physics II, University of Malaga, 29071 Malaga, Spain
| |
Collapse
|
55
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|
56
|
Sell M, Lopes AR, Escudeiro M, Esteves B, Monteiro AR, Trindade T, Cruz-Lopes L. Application of Nanoparticles in Cancer Treatment: A Concise Review. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2887. [PMID: 37947732 PMCID: PMC10650201 DOI: 10.3390/nano13212887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
Timely diagnosis and appropriate antitumoral treatments remain of utmost importance, since cancer remains a leading cause of death worldwide. Within this context, nanotechnology offers specific benefits in terms of cancer therapy by reducing its adverse effects and guiding drugs to selectively target cancer cells. In this comprehensive review, we have summarized the most relevant novel outcomes in the range of 2010-2023, covering the design and application of nanosystems for cancer therapy. We have established the general requirements for nanoparticles to be used in drug delivery and strategies for their uptake in tumor microenvironment and vasculature, including the reticuloendothelial system uptake and surface functionalization with protein corona. After a brief review of the classes of nanovectors, we have covered different classes of nanoparticles used in cancer therapies. First, the advances in the encapsulation of drugs (such as paclitaxel and fisetin) into nanoliposomes and nanoemulsions are described, as well as their relevance in current clinical trials. Then, polymeric nanoparticles are presented, namely the ones comprising poly lactic-co-glycolic acid, polyethylene glycol (and PEG dilemma) and dendrimers. The relevance of quantum dots in bioimaging is also covered, namely the systems with zinc sulfide and indium phosphide. Afterwards, we have reviewed gold nanoparticles (spheres and anisotropic) and their application in plasmon-induced photothermal therapy. The clinical relevance of iron oxide nanoparticles, such as magnetite and maghemite, has been analyzed in different fields, namely for magnetic resonance imaging, immunotherapy, hyperthermia, and drug delivery. Lastly, we have covered the recent advances in the systems using carbon nanomaterials, namely graphene oxide, carbon nanotubes, fullerenes, and carbon dots. Finally, we have compared the strategies of passive and active targeting of nanoparticles and their relevance in cancer theranostics. This review aims to be a (nano)mark on the ongoing journey towards realizing the remarkable potential of different nanoparticles in the realm of cancer therapeutics.
Collapse
Affiliation(s)
- Mariana Sell
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
| | - Ana Rita Lopes
- Faculty of Dental Medicine, Portuguese Catholic University, 3504-505 Viseu, Portugal;
| | - Maria Escudeiro
- Abel Salazar Biomedical Institute, University of Porto, 4050-313 Porto, Portugal;
| | - Bruno Esteves
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
- Centre for Natural Resources, Environment and Society-CERNAS-IPV Research Centre, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal
| | - Ana R. Monteiro
- Centro de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15705 Santiago de Compostela, Spain;
| | - Tito Trindade
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Luísa Cruz-Lopes
- Polytechnic Institute of Viseu, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal; (M.S.); (B.E.)
- Centre for Natural Resources, Environment and Society-CERNAS-IPV Research Centre, Av. Cor. José Maria Vale de Andrade, 3504-510 Viseu, Portugal
| |
Collapse
|
57
|
Wang W, Tasset A, Pyatnitskiy I, Lin P, Bellamkonda A, Mehta R, Gabbert C, Yuan F, Mohamed HG, Peppas NA, Wang H. Reversible, Covalent DNA Condensation Approach Using Chemical Linkers for Enhanced Gene Delivery. NANO LETTERS 2023; 23:9310-9318. [PMID: 37843021 DOI: 10.1021/acs.nanolett.3c02429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Nonviral gene delivery has emerged as a promising technology for gene therapy. Nonetheless, these approaches often face challenges, primarily associated with lower efficiency, which can be attributed to the inefficient transportation of DNA into the nucleus. Here, we report a two-stage condensation approach to achieve efficient nuclear transport of DNA. First, we utilize chemical linkers to cross-link DNA plasmids via a reversible covalent bond to form smaller-sized bundled DNA (b-DNA). Then, we package the b-DNA into cationic vectors to further condense b-DNA and enable efficient gene delivery to the nucleus. We demonstrate clear improvements in the gene transfection efficiency in vitro, including with 11.6 kbp plasmids and in primary cultured neurons. Moreover, we also observed a remarkable improvement in lung-selective gene transfection efficiency in vivo by this two-stage condensation approach following intravenous administration. This reversible covalent assembly strategy demonstrates substantial value of nonviral gene delivery for clinical therapeutic applications.
Collapse
Affiliation(s)
- Wenliang Wang
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Aaron Tasset
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Ilya Pyatnitskiy
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Peter Lin
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Arjun Bellamkonda
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Rohan Mehta
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Christian Gabbert
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Feng Yuan
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Heba Galaa Mohamed
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Nicholas A Peppas
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, Texas 78712, United States
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Huiliang Wang
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
58
|
Lustig DR, Buz E, Mulvey JT, Patterson JP, Kittilstved KR, Sambur JB. Characterizing the Ligand Shell Morphology of PEG-Coated ZnO Nanocrystals Using FRET Spectroscopy. J Phys Chem B 2023; 127:8961-8973. [PMID: 37802098 DOI: 10.1021/acs.jpcb.3c04900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
Poly(ethylene glycol) (PEG) ligands can inhibit proteins and other biomolecules from adhering to underlying surfaces, making them excellent surface ligands for nanocrystal (NC)-based drug carriers. Quantifying the PEG ligand shell morphology is important because its structure determines the permeability of biomolecules through the shell to the NC surface. However, few in situ analytical tools can reveal whether the PEG ligands form either an impenetrable barrier or a porous coating surrounding the NC. Here, we present a Förster resonance energy transfer (FRET) spectroscopy-based approach that can assess the permeability of molecules through PEG-coated ZnO NCs. In this approach, ZnO NCs serve as FRET donors, and freely diffusing molecules in the bulk solution are FRET acceptors. We synthesized a series of variable chain length PEG-silane-coated ZnO NCs such that the longest chain length ligands far exceed the Förster radius (R0), where the energy transfer (EnT) efficiency is 50%. We quantified the EnT efficiency as a function of the ligand chain length using time-resolved photoluminescence lifetime (TRPL) spectroscopy within the framework of FRET theory. Unexpectedly, the longest PEG-silane ligand showed equivalent EnT efficiency as that of bare, hydroxyl-passivated ZnO NCs. These results indicate that the "rigid shell" model fails and the PEG ligand shell morphology is more likely porous or in a patchy "mushroom state", consistent with transmission electron microscopy data. While the spectroscopic measurements and data analysis procedures discussed herein cannot directly visualize the ligand shell morphology in real space, the in situ spectroscopy approach can provide researchers with valuable information regarding the permeability of species through the ligand shell under practical biological conditions.
Collapse
Affiliation(s)
- Danielle R Lustig
- Department of Chemistry, Colorado State University, 200 West Lake Street, Fort Collins, Colorado 80523-1872, United States
| | - Enes Buz
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Justin T Mulvey
- Center for Complex and Active Materials, University of California, Irvine, Irvine, California 92697-2025, United States
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697-2025, United States
| | - Joseph P Patterson
- Center for Complex and Active Materials, University of California, Irvine, Irvine, California 92697-2025, United States
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, United States
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697-2025, United States
| | - Kevin R Kittilstved
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Justin B Sambur
- Department of Chemistry, Colorado State University, 200 West Lake Street, Fort Collins, Colorado 80523-1872, United States
| |
Collapse
|
59
|
Jung S, Ben Nasr M, Bahmani B, Usuelli V, Zhao J, Sabiu G, Seelam AJ, Naini SM, Balasubramanian HB, Park Y, Li X, Khalefa SA, Kasinath V, Williams MD, Rachid O, Haik Y, Tsokos GC, Wasserfall CH, Atkinson MA, Bromberg JS, Tao W, Fiorina P, Abdi R. Nanotargeted Delivery of Immune Therapeutics in Type 1 Diabetes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300812. [PMID: 37357903 PMCID: PMC10629472 DOI: 10.1002/adma.202300812] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/14/2023] [Indexed: 06/27/2023]
Abstract
Immune therapeutics holds great promise in the treatment of type 1 diabetes (T1D). Nonetheless, their progress is hampered by limited efficacy, equipoise, or issues of safety. To address this, a novel and specific nanodelivery platform for T1D that targets high endothelial venules (HEVs) presented in the pancreatic lymph nodes (PLNs) and pancreas is developed. Data indicate that the pancreata of nonobese diabetic (NOD) mice and patients with T1D are unique in their expression of newly formed HEVs. Anti-CD3 mAb is encapsulated in poly(lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles (NPs), the surfaces of which are conjugated with MECA79 mAb that recognizes HEVs. Targeted delivery of these NPs improves accumulation of anti-CD3 mAb in both the PLNs and pancreata of NOD mice. Treatment of hyperglycemic NOD mice with MECA79-anti-CD3-NPs results in significant reversal of T1D compared to those that are untreated, treated with empty NPs, or provided free anti-CD3. This effect is associated with a significant reduction of T effector cell populations in the PLNs and a decreased production of pro-inflammatory cytokine in the mice treated with MECA79-anti-CD3-NPs. In summary, HEV-targeted therapeutics may be used as a means by which immune therapeutics can be delivered to PLNs and pancreata to suppress autoimmune diabetes effectively.
Collapse
Affiliation(s)
- Sungwook Jung
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Moufida Ben Nasr
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
| | - Baharak Bahmani
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Vera Usuelli
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
| | - Jing Zhao
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gianmarco Sabiu
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Andy Joe Seelam
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Said Movahedi Naini
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hari Baskar Balasubramanian
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
| | - Youngrong Park
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaofei Li
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Salma Ayman Khalefa
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
| | - Vivek Kasinath
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - MacKenzie D Williams
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Ousama Rachid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, 2713, Doha, Qatar
| | - Yousef Haik
- Department of Mechanical and Nuclear Engineering, University of Sharjah, 27272, Sharjah, UAE
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
- Department of Pediatrics, University of Florida, Gainesville, FL, 32610, USA
| | - Jonathan S Bromberg
- Departments of Surgery and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wei Tao
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Paolo Fiorina
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Reza Abdi
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
60
|
Stengel D, Demirel BH, Knoll P, Truszkowska M, Laffleur F, Bernkop-Schnürch A. PEG vs. zwitterions: How these surface decorations determine cellular uptake of lipid-based nanocarriers. J Colloid Interface Sci 2023; 647:52-64. [PMID: 37244176 DOI: 10.1016/j.jcis.2023.05.079] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/24/2023] [Accepted: 05/13/2023] [Indexed: 05/29/2023]
Abstract
AIM To evaluate the impact of polyethylene glycol (PEG) and zwitterionic surface decoration of lipid-based nanocarriers (NC) on cellular uptake. METHODS Anionic, neutral and cationic zwitterionic lipid-based NCs based on lecithin were compared with conventional PEGylated lipid-based NCs regarding stability in biorelevant fluids, interaction with endosome mimicking membranes, cytocompatibility, cellular uptake and permeation across intestinal mucosa. RESULTS PEGylated and zwitterionic lipid-based NCs exhibited a droplet size between 100 and 125 nm with a narrow size distribution. For the PEGylated and zwitterionic lipid-based NCs only minor alterations in size and PDI in fasted state intestinal fluid and mucus containing buffer were observed, demonstrating similar bioinert properties. Erythrocytes interaction studies revealed enhanced endosomal escape properties for zwitterionic lipid-based NCs compared to PEGylated lipid-based NCs. For the zwitterionic lipid-based NCs negligible cytotoxicity on Caco-2 and HEK cells, even in the highest tested concentration of 1 % (v/v) was recorded. The PEGylated lipid-based NCs showed a cell survival of ≥75 % for concentrations ≤0.05 % on Caco-2 and HEK cells, which was considered as non-toxic. For the zwitterionic lipid-based NCs up to 60-fold higher cellular uptake on Caco-2 cells was determined compared to PEGylated lipid-based NCs. For the cationic zwitterionic lipid-based NCs the highest cellular uptake with 58.5 % and 40.0 % in Caco-2 and HEK cells, respectively, was determined. The results were confirmed visually by life cell imaging. Ex-vivo permeation experiments using rat intestinal mucosa demonstrated up to 8.6-fold enhanced permeation of the lipophilic marker coumarin-6 in zwitterionic lipid-based NCs compared to the control. Up to 6.9-fold enhanced permeation of coumarin-6 in neutral zwitterionic lipid-based NCs compared to the PEGylated counterpart was recorded. CONCLUSION The replacement of PEG surfactants with zwitterionic surfactants is a promising approach to overcome the drawbacks of conventional PEGylated lipid-based NCs regarding intracellular drug delivery.
Collapse
Affiliation(s)
- Daniel Stengel
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Betül Hilal Demirel
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Patrick Knoll
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Martyna Truszkowska
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Flavia Laffleur
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria.
| |
Collapse
|
61
|
Lebreton V, Legeay S, Vasylaki A, Lagarce F, Saulnier P. Protein corona formation on lipidic nanocapsules: Influence of the interfacial PEG repartition. Eur J Pharm Sci 2023; 189:106537. [PMID: 37490974 DOI: 10.1016/j.ejps.2023.106537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/04/2023] [Accepted: 07/22/2023] [Indexed: 07/27/2023]
Abstract
The parameters currently used for characterization of nanoparticles, such as size and zeta potential, were not able to reflect the performance of a nanocarrier in the biological environment. Therefore, more thorough in vitro characterization is required to predict their behavior in vivo, where nanoparticles acquire a new biological identity due to interactions with biomolecules. In this present study, we performed in vitro characterization in biological fluids for lipid nanocapsules (LNCs) with varying means sizes (50 nm and 100 nm), different electrical surface charges and different Poly Ethylene Glycol (PEG) compositions. Then, different methods were applied to show the impact of the protein corona formation on LNCs. Even if all formulations attached to plasmatic proteins, a higher thickness of corona and highest protein binding was observed for certain LNC50 formulations. A better knowledge of the phenomenon of protein adsorption over NPs in the plasmatic media is a cornerstone of clinical translation. In fact, after short blood circulation time, it is not the initially designed nanoparticle but the complex nanoparticle bearing its protein corona which circulates to reach its target.
Collapse
Affiliation(s)
- Vincent Lebreton
- MINT, UNIV Angers, SFR-ICAT, INSERM U1066, CNRS 6021, Angers 4208, France; CHU Angers, Angers 49033, France.
| | - Samuel Legeay
- MINT, UNIV Angers, SFR-ICAT, INSERM U1066, CNRS 6021, Angers 4208, France
| | | | - Fredéric Lagarce
- MINT, UNIV Angers, SFR-ICAT, INSERM U1066, CNRS 6021, Angers 4208, France; CHU Angers, Angers 49033, France
| | - Patrick Saulnier
- MINT, UNIV Angers, SFR-ICAT, INSERM U1066, CNRS 6021, Angers 4208, France; CHU Angers, Angers 49033, France
| |
Collapse
|
62
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
63
|
Schmid R, Kaiser J, Willbold R, Walther N, Wittig R, Lindén M. Towards a simple in vitro surface chemistry pre-screening method for nanoparticles to be used for drug delivery to solid tumours. Biomater Sci 2023; 11:6287-6298. [PMID: 37551433 DOI: 10.1039/d3bm00966a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
An efficient nanoparticulate drug carrier intended for chemotherapy based on intravenous administration must exhibit a long enough blood circulation time, a good penetrability into the tumour volume, as well as an efficient uptake by cancer cells. Limiting factors for the therapeutic outcome in vivo are recognition of the nanoparticles as foreign objects, which triggers nanoparticle uptake by defence organs rich in macrophages, e.g. liver and spleen, on the time-scale of accumulation and uptake in/by the tumour. However, the development of nanomedicine towards efficient nanoparticle-based delivery to solid tumours is hampered by the lack of simple, reproducible, cheap, and predictive means for early identification of promising nanoparticle formulations. The surface chemistry of nanoparticles is known to be the most important determinant for the biological fate of nanoparticles, as it influences the extent of serum protein adsorption, and also the relative composition of the protein corona. Here we preliminarily evaluate an extremely simple screening method for nanoparticle surface chemistry pre-optimization based on nanoparticle uptake in vitro by PC-3 cancer cells and THP-1 macrophages. Only when both selectivity for the cancer cells as well as the extent of nanoparticle uptake are taken into consideration do the in vitro results mirror literature results obtained for small animal models. Furthermore, although not investigated here, the screening method does also lend itself to the study of actively targeted nanoparticles.
Collapse
Affiliation(s)
- Roman Schmid
- Inorganic Chemistry II, Albert-Einstein-Allee 11, Ulm University, 89081 Ulm, Germany.
| | - Juliane Kaiser
- Institute for Laser Technologies in Medicine & Metrology (ILM) at Ulm University, Helmholtzstrasse 12, 89081 Ulm, Germany.
| | - Ramona Willbold
- Institute for Laser Technologies in Medicine & Metrology (ILM) at Ulm University, Helmholtzstrasse 12, 89081 Ulm, Germany.
| | - Nomusa Walther
- Institute for Laser Technologies in Medicine & Metrology (ILM) at Ulm University, Helmholtzstrasse 12, 89081 Ulm, Germany.
| | - Rainer Wittig
- Institute for Laser Technologies in Medicine & Metrology (ILM) at Ulm University, Helmholtzstrasse 12, 89081 Ulm, Germany.
| | - Mika Lindén
- Inorganic Chemistry II, Albert-Einstein-Allee 11, Ulm University, 89081 Ulm, Germany.
| |
Collapse
|
64
|
Gao J, Stengel P, Lu T, Wu Y, Hawker DD, Gutowski KE, Hankett JM, Kellermeier M, Chen Z. Antiadhesive Copolymers at Solid/Liquid Interfaces: Complementary Characterization of Polymer Adsorption and Protein Fouling by Sum Frequency Generation Vibrational Spectroscopy and Quartz-Crystal Microbalance Measurements with Dissipation Monitoring. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:12270-12282. [PMID: 37586045 DOI: 10.1021/acs.langmuir.3c01759] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Amphiphilic copolymers comprising hydrophilic segments of poly(ethylene glycol) and hydrophobic domains that are able to adhere to solid/liquid interfaces have proven to be versatile ingredients in formulated products for various types of applications. Recently, we have reported the successful synthesis of a copolymer designed for modifying the surface properties of polyesters as mimics for synthetic textiles. Using sum frequency generation (SFG) spectroscopy, it was shown that the newly developed copolymer adsorbs effectively on the targeted substrates even in the presence of surfactants as supplied by common detergents. In the present work, these studies were extended to evaluate the ability of the formed copolymer adlayers to passivate polyester surfaces against undesired deposition of bio(macro)molecules, as represented by fibrinogen as model protein foulants. In addition, SFG spectroscopy was used to elucidate the structure of fibrinogen at the interface between polyester and water. To complement the obtained data with an independent technique, analogous experiments were performed using quartz-crystal microbalance with dissipation monitoring for the detection of the relevant interfacial processes. Both methods give consistent results and deliver a holistic picture of brush copolymer adsorption on polyester surfaces and subsequent antiadhesive effects against proteins under different conditions representing the targeted application in home care products.
Collapse
Affiliation(s)
- Jinpeng Gao
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Peter Stengel
- Material Science, BASF SE, RGA/BM - B007, Carl-Bosch-Strasse 38, D-67056 Ludwigshafen, Germany
| | - Tieyi Lu
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Yuchen Wu
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
- Department of Macromolecular Science and Engineering, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Dustin D Hawker
- BASF Corporation, 1609 Biddle Avenue, Wyandotte, Michigan 48192, United States
| | - Keith E Gutowski
- BASF Corporation, 1609 Biddle Avenue, Wyandotte, Michigan 48192, United States
| | - Jeanne M Hankett
- BASF Corporation, 1609 Biddle Avenue, Wyandotte, Michigan 48192, United States
| | - Matthias Kellermeier
- Material Science, BASF SE, RGA/BM - B007, Carl-Bosch-Strasse 38, D-67056 Ludwigshafen, Germany
| | - Zhan Chen
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
- Department of Macromolecular Science and Engineering, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
65
|
Subbotina J, Rouse I, Lobaskin V. In silico prediction of protein binding affinities onto core-shell PEGylated noble metal nanoparticles for rational design of drug nanocarriers. NANOSCALE 2023; 15:13371-13383. [PMID: 37530535 DOI: 10.1039/d3nr03264g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Polymer-coated nanoparticles (NP) are commonly used as drug carriers or theranostic agents. Their uptake rates are modulated by the interactions with essential serum proteins such as transferrin and albumin. Understanding the control parameters of these interactions is crucial for improving the efficiency of these nanoscale devices. In this work, we perform a multiscale computational study of protein adsorption onto polyethylene glycol (PEG) coated gold and silver NPs, producing protein-NP adsorption rankings as a function of PEG grafting density, which are validated against previously reported experimental protein-NP binding constants. Furthermore, the applied nano-docking method provides information on the preferred orientation of proteins immobilised on the surface of NPs. We propose a method of construction of model core-shell NPs in silico. The presented protocol can provide molecular level insights for the experimental development of biosensors, nanocarriers, or other nanoplatforms where information on the preferred orientation of protein at the bio-nano interface is crucial, and enables fast in silico prescreening of assays of various nanocarriers, i.e., combinations of proteins, NPs, and coatings.
Collapse
Affiliation(s)
- Julia Subbotina
- School of Physics, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Ian Rouse
- School of Physics, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Vladimir Lobaskin
- School of Physics, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
66
|
Polyak A, Harting H, Angrisani N, Herrmann T, Ehlert N, Meißner J, Willmann M, Al-Bazaz S, Ross TL, Bankstahl JP, Reifenrath J. Preparation and PET/CT imaging of implant directed 68Ga-labeled magnetic nanoporous silica nanoparticles. J Nanobiotechnology 2023; 21:270. [PMID: 37592318 PMCID: PMC10433681 DOI: 10.1186/s12951-023-02041-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Implant infections caused by biofilm forming bacteria are a major threat in orthopedic surgery. Delivering antibiotics directly to an implant affected by a bacterial biofilm via superparamagnetic nanoporous silica nanoparticles could present a promising approach. Nevertheless, short blood circulation half-life because of rapid interactions of nanoparticles with the host's immune system hinder them from being clinically used. The aim of this study was to determine the temporal in vivo resolution of magnetic nanoporous silica nanoparticle (MNPSNP) distribution and the effect of PEGylation and clodronate application using PET/CT imaging and gamma counting in an implant mouse model. METHODS PEGylated and non-PEGylated MNPSNPs were radiolabeled with gallium-68 (68Ga), implementing the chelator tris(hydroxypyridinone). 36 mice were included in the study, 24 mice received a magnetic implant subcutaneously on the left and a titanium implant on the right hind leg. MNPSNP pharmacokinetics and implant accumulation was analyzed in dependence on PEGylation and additional clodronate application. Subsequently gamma counting was performed for further final analysis. RESULTS The pharmacokinetics and biodistribution of all radiolabeled nanoparticles could clearly be visualized and followed by dynamic PET/CT imaging. Both variants of 68Ga-labeled MNPSNP accumulated mainly in liver and spleen. PEGylation of the nanoparticles already resulted in lower liver uptakes. Combination with macrophage depletion led to a highly significant effect whereas macrophage depletion alone could not reveal significant differences. Although MNPSNP accumulation around implants was low in comparison to the inner organs in PET/CT imaging, gamma counting displayed a significantly higher %I.D./g for the tissue surrounding the magnetic implants compared to the titanium control. Additional PEGylation and/or macrophage depletion revealed no significant differences regarding nanoparticle accumulation at the implantation site. CONCLUSION Tracking of 68Ga-labeled nanoparticles in a mouse model in the first critical hours post-injection by PET/CT imaging provided a better understanding of MNPSNP distribution, elimination and accumulation. Although PEGylation increases circulation time, nanoparticle accumulation at the implantation site was still insufficient for infection treatment and additional efforts are needed to increase local accumulation.
Collapse
Affiliation(s)
- Andras Polyak
- NIFE - Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Heidi Harting
- NIFE - Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany.
- Clinic for Orthopedic Surgery, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Nina Angrisani
- NIFE - Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
- Clinic for Orthopedic Surgery, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Timo Herrmann
- Institute for Inorganic Chemistry, Leibniz University Hannover, Callinstraße 9, 30167, Hannover, Germany
| | - Nina Ehlert
- Institute for Inorganic Chemistry, Leibniz University Hannover, Callinstraße 9, 30167, Hannover, Germany
| | - Jessica Meißner
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hanover, Foundation, Buenteweg 17, 30559, Hannover, Germany
| | - Michael Willmann
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Silav Al-Bazaz
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Janin Reifenrath
- NIFE - Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625, Hannover, Germany
- Clinic for Orthopedic Surgery, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| |
Collapse
|
67
|
Park S, Ha MK, Lee Y, Song J, Yoon TH. Effects of Immune Cell Heterogeneity and Protein Corona on the Cellular Association and Cytotoxicity of Gold Nanoparticles: A Single-Cell-Based, High-Dimensional Mass Cytometry Study. ACS NANOSCIENCE AU 2023; 3:323-334. [PMID: 37601916 PMCID: PMC10436372 DOI: 10.1021/acsnanoscienceau.3c00001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 08/22/2023]
Abstract
Understanding how nanoparticles (NPs) interact with biological systems is important in many biomedical research areas. However, the heterogeneous nature of biological systems, including the existence of numerous cell types and multitudes of key environmental factors, makes these interactions extremely challenging to investigate precisely. Here, using a single-cell-based, high-dimensional mass cytometry approach, we demonstrated that the presence of protein corona has significant influences on the cellular associations and cytotoxicity of gold NPs for human immune cells, and those effects vary significantly with the types of immune cells and their subsets. The altered surface functionality of protein corona reduced the cytotoxicity and cellular association of gold NPs in most cell types (e.g., monocytes, dendritic cells, B cells, natural killer (NK) cells, and T cells) and those immune cells selected different endocytosis pathways such as receptor-mediated endocytosis, phagocytosis, and micropinocytosis. However, even slight alterations in the major cell type (phagocytic cells and non-phagocytic cells) and T cell subsets (e.g., memory and naive T cells) resulted in significant protein corona-dependent variations in their cellular dose of gold NPs. Especially, naive T killer cells exhibited additional heterogeneity than memory T killer cells, with clusters exhibiting distinct cellular association patterns in single-cell contour plots. This multi-parametric analysis of mass cytometry data established a conceptual framework for a more holistic understanding of how the human immune system responds to external stimuli, paving the way for the application of precisely engineered NPs as promising tools of nanomedicine under various clinical settings, including targeted drug delivery and vaccine development.
Collapse
Affiliation(s)
- Sehee Park
- Department
of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Republic
of Korea
| | - My Kieu Ha
- Department
of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Republic
of Korea
| | - Yangsoon Lee
- Department
of Laboratory Medicine, College of Medicine, Hanyang University, Seoul 04763, Republic
of Korea
| | - Jaewoo Song
- Department
of Laboratory Medicine, College of Medicine, Yonsei University, Seoul 03722, Republic
of Korea
| | - Tae Hyun Yoon
- Department
of Chemistry, College of Natural Sciences, Hanyang University, Seoul 04763, Republic
of Korea
- Research
Institute for Convergence of Basic Science, Hanyang University, Seoul 04763, Republic
of Korea
- Institute
of Next Generation Material Design, Hanyang
University, Seoul 04763, Republic of Korea
- Yoon
Idea
Lab. Co. Ltd, Seoul 04763, Republic of Korea
| |
Collapse
|
68
|
Alexander S, Moghadam MG, Rothenbroker M, Y T Chou L. Addressing the in vivo delivery of nucleic-acid nanostructure therapeutics. Adv Drug Deliv Rev 2023; 199:114898. [PMID: 37230305 DOI: 10.1016/j.addr.2023.114898] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
DNA and RNA nanostructures are being investigated as therapeutics, vaccines, and drug delivery systems. These nanostructures can be functionalized with guests ranging from small molecules to proteins with precise spatial and stoichiometric control. This has enabled new strategies to manipulate drug activity and to engineer devices with novel therapeutic functionalities. Although existing studies have offered encouraging in vitro or pre-clinical proof-of-concepts, establishing mechanisms of in vivo delivery is the new frontier for nucleic-acid nanotechnologies. In this review, we first provide a summary of existing literature on the in vivo uses of DNA and RNA nanostructures. Based on their application areas, we discuss current models of nanoparticle delivery, and thereby highlight knowledge gaps on the in vivo interactions of nucleic-acid nanostructures. Finally, we describe techniques and strategies for investigating and engineering these interactions. Together, we propose a framework to establish in vivo design principles and advance the in vivo translation of nucleic-acid nanotechnologies.
Collapse
Affiliation(s)
- Shana Alexander
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | | | - Meghan Rothenbroker
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Leo Y T Chou
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada.
| |
Collapse
|
69
|
Wang X, Zhang H, Chen X, Wu C, Ding K, Sun G, Luo Y, Xiang D. Overcoming tumor microenvironment obstacles: Current approaches for boosting nanodrug delivery. Acta Biomater 2023; 166:42-68. [PMID: 37257574 DOI: 10.1016/j.actbio.2023.05.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
In order to achieve targeted delivery of anticancer drugs, efficacy improvement, and side effect reduction, various types of nanoparticles are employed. However, their therapeutic effects are not ideal. This phenomenon is caused by tumor microenvironment abnormalities such as abnormal blood vessels, elevated interstitial fluid pressure, and dense extracellular matrix that affect nanoparticle penetration into the tumor's interstitium. Furthermore, nanoparticle properties including size, charge, and shape affect nanoparticle transport into tumors. This review comprehensively goes over the factors hindering nanoparticle penetration into tumors and describes methods for improving nanoparticle distribution by remodeling the tumor microenvironment and optimizing nanoparticle physicochemical properties. Finally, a critical analysis of future development of nanodrug delivery in oncology is further discussed. STATEMENT OF SIGNIFICANCE: This article reviews the factors that hinder the distribution of nanoparticles in tumors, and describes existing methods and approaches for improving the tumor accumulation from the aspects of remodeling the tumor microenvironment and optimizing the properties of nanoparticles. The description of the existing methods and approaches is followed by highlighting their advantages and disadvantages and put forward possible directions for the future researches. At last, the challenges of improving tumor accumulation in nanomedicines design were also discussed. This review will be of great interest to the broad readers who are committed to delivering nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Xiaohui Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Hong Zhang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Xiaohui Chen
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chunrong Wu
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Ke Ding
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Guiyin Sun
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Debing Xiang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| |
Collapse
|
70
|
Wang S, Zhang J, Zhou H, Lu YC, Jin X, Luo L, You J. The role of protein corona on nanodrugs for organ-targeting and its prospects of application. J Control Release 2023; 360:15-43. [PMID: 37328008 DOI: 10.1016/j.jconrel.2023.06.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/30/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023]
Abstract
Nowadays, nanodrugs become a hotspot in the high-end medical field. They have the ability to deliver drugs to reach their destination more effectively due to their unique properties and flexible functionalization. However, the fate of nanodrugs in vivo is not the same as those presented in vitro, which indeed influenced their therapeutic efficacy in vivo. When entering the biological organism, nanodrugs will first come into contact with biological fluids and then be covered by some biomacromolecules, especially proteins. The proteins adsorbed on the surface of nanodrugs are known as protein corona (PC), which causes the loss of prospective organ-targeting abilities. Fortunately, the reasonable utilization of PC may determine the organ-targeting efficiency of systemically administered nanodrugs based on the diverse expression of receptors on cells in different organs. In addition, the nanodrugs for local administration targeting diverse lesion sites will also form unique PC, which plays an important role in the therapeutic effect of nanodrugs. This article introduced the formation of PC on the surface of nanodrugs and summarized the recent studies about the roles of diversified proteins adsorbed on nanodrugs and relevant protein for organ-targeting receptor through different administration pathways, which may deepen our understanding of the role that PC played on organ-targeting and improve the therapeutic efficacy of nanodrugs to promote their clinical translation.
Collapse
Affiliation(s)
- Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Junlei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Huanli Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Yi Chao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Xizhi Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China; Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou 310058, PR China; Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China.
| |
Collapse
|
71
|
Shafaei N, Khorshidi S, Karkhaneh A. The immune-stealth polymeric coating on drug delivery nanocarriers: In vitro engineering and in vivo fate. J Biomater Appl 2023:8853282231185352. [PMID: 37480331 DOI: 10.1177/08853282231185352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Although essential nanosystems such as nanoparticles and nanocarriers are desirable options for transporting various drug molecules into the biological environment, they rapidly remove from the circulatory system due to their interaction with multiple in vivo barriers, especially the immune barrier, which will result in their short-term effects. In order to improve their effectiveness and durability in the circulatory system, the polymer coatings can use to cover the surface of nanoparticles and nanocarriers to conceal them from the immune system. Due to their different properties (like charge, elasticity, and hydrophilicity/hydrophobicity), these coatings can improve drug delivery nanosystem durability and therapeutic applications. The mentioned coatings have different types and are divided into various categories, such as synthetic polymers, polysaccharides, and zwitterionic polymers. Each of these polymers has unique properties based on its category, origin, and chemical structure that make them suitable for producing stealth drug delivery nanocarriers. In this review article, we have tried to explain the importance of these diverse polymer coatings in determining the fate of drug nanocarriers and then introduced the different types of these coatings and, finally, described various methods that directly and indirectly analyze the nanocoatings to determine the stability of nanoparticles in the body.
Collapse
Affiliation(s)
- Nadia Shafaei
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Sajedeh Khorshidi
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Akbar Karkhaneh
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
72
|
Novorolsky RJ, Kasheke GDS, Hakim A, Foldvari M, Dorighello GG, Sekler I, Vuligonda V, Sanders ME, Renden RB, Wilson JJ, Robertson GS. Preserving and enhancing mitochondrial function after stroke to protect and repair the neurovascular unit: novel opportunities for nanoparticle-based drug delivery. Front Cell Neurosci 2023; 17:1226630. [PMID: 37484823 PMCID: PMC10360135 DOI: 10.3389/fncel.2023.1226630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glia, and neurons that form the basic component of the blood brain barrier. This intricate structure rapidly adjusts cerebral blood flow to match the metabolic needs of brain activity. However, the NVU is exquisitely sensitive to damage and displays limited repair after a stroke. To effectively treat stroke, it is therefore considered crucial to both protect and repair the NVU. Mitochondrial calcium (Ca2+) uptake supports NVU function by buffering Ca2+ and stimulating energy production. However, excessive mitochondrial Ca2+ uptake causes toxic mitochondrial Ca2+ overloading that triggers numerous cell death pathways which destroy the NVU. Mitochondrial damage is one of the earliest pathological events in stroke. Drugs that preserve mitochondrial integrity and function should therefore confer profound NVU protection by blocking the initiation of numerous injury events. We have shown that mitochondrial Ca2+ uptake and efflux in the brain are mediated by the mitochondrial Ca2+ uniporter complex (MCUcx) and sodium/Ca2+/lithium exchanger (NCLX), respectively. Moreover, our recent pharmacological studies have demonstrated that MCUcx inhibition and NCLX activation suppress ischemic and excitotoxic neuronal cell death by blocking mitochondrial Ca2+ overloading. These findings suggest that combining MCUcx inhibition with NCLX activation should markedly protect the NVU. In terms of promoting NVU repair, nuclear hormone receptor activation is a promising approach. Retinoid X receptor (RXR) and thyroid hormone receptor (TR) agonists activate complementary transcriptional programs that stimulate mitochondrial biogenesis, suppress inflammation, and enhance the production of new vascular cells, glia, and neurons. RXR and TR agonism should thus further improve the clinical benefits of MCUcx inhibition and NCLX activation by increasing NVU repair. However, drugs that either inhibit the MCUcx, or stimulate the NCLX, or activate the RXR or TR, suffer from adverse effects caused by undesired actions on healthy tissues. To overcome this problem, we describe the use of nanoparticle drug formulations that preferentially target metabolically compromised and damaged NVUs after an ischemic or hemorrhagic stroke. These nanoparticle-based approaches have the potential to improve clinical safety and efficacy by maximizing drug delivery to diseased NVUs and minimizing drug exposure in healthy brain and peripheral tissues.
Collapse
Affiliation(s)
- Robyn J. Novorolsky
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Gracious D. S. Kasheke
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Antoine Hakim
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Marianna Foldvari
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Gabriel G. Dorighello
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben Gurion University, Beersheva, Israel
| | | | | | - Robert B. Renden
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, College of Arts and Sciences, Cornell University, Ithaca, NY, United States
| | - George S. Robertson
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Department of Psychiatry, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
73
|
Rosini E, Boreggio M, Verga M, Caldinelli L, Pollegioni L, Fasoli E. The D-amino acid oxidase-carbon nanotubes: evaluation of cytotoxicity and biocompatibility of a potential anticancer nanosystem. 3 Biotech 2023; 13:243. [PMID: 37346390 PMCID: PMC10279611 DOI: 10.1007/s13205-023-03568-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/19/2023] [Indexed: 06/23/2023] Open
Abstract
The 'enzyme prodrug therapy' represents a promising strategy to overcome limitations of current cancer treatments by the systemic administration of prodrugs, converted by a foreign enzyme into an active anticancer compound directly in tumor sites. One example is D-amino acid oxidase (DAAO), a dimeric flavoenzyme able to catalyze the oxidative deamination of D-amino acids with production of hydrogen peroxide, a reactive oxygen species (ROS), able to favor cancer cells death. A DAAO variant containing five aminoacidic substitutions (mDAAO) was demonstrated to possess a better therapeutic efficacy under low O2 concentration than wild-type DAAO (wtDAAO). Recently, aiming to design promising nanocarriers for DAAO, multi-walled carbon nanotubes (MWCNTs) were functionalized with polyethylene glycol (PEG) to reduce their tendency to aggregation and to improve their biocompatibility. Here, wtDAAO and mDAAO were adsorbed on PEGylated MWCNTs and their activity and cytotoxicity were tested. While PEG-MWCNTs-DAAOs have shown a higher activity than pristine MWCNTs-DAAO (independently on the DAAO variant used), PEG-MWCNTs-mDAAO showed a higher cytotoxicity than PEG-MWCNTs-wtDAAO at low O2 concentration. In order to evaluate the nanocarriers' biocompatibility, PEG-MWCNTs-DAAOs were incubated in human serum and the composition of protein corona was investigated via nLC-MS/MS, aiming to characterize both soft and hard coronas. The mDAAO variant has influenced the bio-corona composition in both number of proteins and presence of opsonins and dysopsonins: notably, the soft corona of PEG-MWCNTs-mDAAO contained less proteins and was more enriched in proteins able to inhibit the immune response than PEG-MWCNTs-wtDAAO. Considering the obtained results, the PEGylated MWCNTs conjugated with the mDAAO variant seems a promising candidate for a selective antitumor oxidative therapy: under anoxic-like conditions, this novel drug delivery system showed a remarkable cytotoxic effect controlled by the substrate addition, against different tumor cell lines, and a bio-corona composition devoted to prolong its blood circulation time, thus improving the drug's biodistribution. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03568-1.
Collapse
Affiliation(s)
- Elena Rosini
- Department of Biotechnology and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Marta Boreggio
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | - Matteo Verga
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | - Laura Caldinelli
- Department of Biotechnology and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Elisa Fasoli
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| |
Collapse
|
74
|
Waggoner LE, Miyasaki KF, Kwon EJ. Analysis of PEG-lipid anchor length on lipid nanoparticle pharmacokinetics and activity in a mouse model of traumatic brain injury. Biomater Sci 2023; 11:4238-4253. [PMID: 36987922 PMCID: PMC10262813 DOI: 10.1039/d2bm01846b] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023]
Abstract
Traumatic brain injury (TBI) affects millions of people worldwide, yet there are currently no therapeutics that address the long-term impairments that develop in a large portion of survivors. Lipid nanoparticles (LNPs) are a promising therapeutic strategy that may address the molecular basis of TBI pathophysiology. LNPs are the only non-viral gene delivery platform to achieve clinical success, but systemically administered formulations have only been established for targets in the liver. In this work, we evaluated the pharmacokinetics and activity of LNPs formulated with polyethylene glycol (PEG)-lipids of different anchor lengths when systemically administered to a mouse model of TBI. We observed an increase in LNP accumulation and activity in the injured brain hemisphere compared to the uninjured contralateral brain hemisphere. Interestingly, transgene expression mediated by LNPs was more durable in injured brain tissue compared to off-target organs when compared between 4 and 24 hours. The PEG-lipid is an important component of LNP formulation necessary for the stable formation and storage of LNPs, but the PEG-lipid structure and content also has an impact on LNP function. LNP formulations containing various ratios of PEG-lipid with C18 (DSPE-PEG) and C14 (DMG-PEG) anchors displayed similar physicochemical properties, independent of the PEG-lipid compositions. As the proportion of DSPE-PEG was increased in formulations, blood circulation times of LNPs increased and the duration of expression increased. We also evaluated diffusion of LNPs after convection enhanced delivery (CED) in healthy brains and found LNPs distributed >1 mm away from the injection site. Understanding LNP pharmacokinetics and activity in TBI models and the impact of PEG-lipid anchor length informs the design of LNP-based therapies for TBI after systemic administration.
Collapse
Affiliation(s)
- Lauren E Waggoner
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Katelyn F Miyasaki
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Ester J Kwon
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
75
|
Tenchov R, Sasso JM, Zhou QA. PEGylated Lipid Nanoparticle Formulations: Immunological Safety and Efficiency Perspective. Bioconjug Chem 2023. [PMID: 37162501 DOI: 10.1021/acs.bioconjchem.3c00174] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Lipid nanoparticles (LNPs) have been recognized as efficient vehicles to transport a large variety of therapeutics. Currently in the spotlight as important constituents of the COVID-19 mRNA vaccines, LNPs play a significant role in protecting and transporting mRNA to cells. As one of their key constituents, polyethylene glycol (PEG)-lipid conjugates are important in defining LNP physicochemical characteristics and biological activity. PEGylation has proven particularly efficient in conferring longer systemic circulation of LNPs, thus greatly improving their pharmacokinetics and efficiency. Along with revealing the benefits of PEG conjugates, studies have revealed unexpected immune reactions against PEGylated nanocarriers such as accelerated blood clearance (ABC), involving the production of anti-PEG antibodies at initial injection, which initiates accelerated blood clearance upon subsequent injections, as well as a hypersensitivity reaction referred to as complement activation-related pseudoallergy (CARPA). Further, data have been accumulated indicating consistent yet sometimes controversial correlations between various structural parameters of the PEG-lipids, the properties of the PEGylated LNPs, and the magnitude of the observed adverse effects. Detailed knowledge and comprehension of such correlations are of foremost importance in the efforts to diminish and eliminate the undesirable immune reactions and improve the safety and efficiency of the PEGylated medicines. Here, we present an overview based on analysis of data from the CAS Content Collection regarding the PEGylated LNP immunogenicity and overall safety concerns. A comprehensive summary has been compiled outlining how various structural parameters of the PEG-lipids affect the immune responses and activities of the LNPs, with regards to their efficiency in drug delivery. This Review is thus intended to serve as a helpful resource in understanding the current knowledge in the field, in an effort to further solve the remaining challenges and to achieve full potential.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M Sasso
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
76
|
Nienhaus K, Nienhaus GU. Mechanistic Understanding of Protein Corona Formation around Nanoparticles: Old Puzzles and New Insights. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2301663. [PMID: 37010040 DOI: 10.1002/smll.202301663] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/14/2023] [Indexed: 06/19/2023]
Abstract
Although a wide variety of nanoparticles (NPs) have been engineered for use as disease markers or drug delivery agents, the number of nanomedicines in clinical use has hitherto remained small. A key obstacle in nanomedicine development is the lack of a deep mechanistic understanding of NP interactions in the bio-environment. Here, the focus is on the biomolecular adsorption layer (protein corona), which quickly enshrouds a pristine NP exposed to a biofluid and modifies the way the NP interacts with the bio-environment. After a brief introduction of NPs for nanomedicine, proteins, and their mutual interactions, research aimed at addressing fundamental properties of the protein corona, specifically its mono-/multilayer structure, reversibility and irreversibility, time dependence, as well as its role in NP agglomeration, is critically reviewed. It becomes quite evident that the knowledge of the protein corona is still fragmented, and conflicting results on fundamental issues call for further mechanistic studies. The article concludes with a discussion of future research directions that should be taken to advance the understanding of the protein corona around NPs. This knowledge will provide NP developers with the predictive power to account for these interactions in the design of efficacious nanomedicines.
Collapse
Affiliation(s)
- Karin Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology, 76049, Karlsruhe, Germany
| | - Gerd Ulrich Nienhaus
- Institute of Applied Physics, Karlsruhe Institute of Technology, 76049, Karlsruhe, Germany
- Institute of Nanotechnology, Karlsruhe Institute of Technology, 76021, Karlsruhe, Germany
- Institute of Biological and Chemical Systems, Karlsruhe Institute of Technology, 76021, Karlsruhe, Germany
- Department of Physics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
77
|
Fulton MD, Najahi-Missaoui W. Liposomes in Cancer Therapy: How Did We Start and Where Are We Now. Int J Mol Sci 2023; 24:ijms24076615. [PMID: 37047585 PMCID: PMC10095497 DOI: 10.3390/ijms24076615] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Since their first discovery in the 1960s by Alec Bangham, liposomes have been shown to be effective drug delivery systems for treating various cancers. Several liposome-based formulations received approval by the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA), with many others in clinical trials. Liposomes have several advantages, including improved pharmacokinetic properties of the encapsulated drug, reduced systemic toxicity, extended circulation time, and targeted disposition in tumor sites due to the enhanced permeability and retention (EPR) mechanism. However, it is worth noting that despite their efficacy in treating various cancers, liposomes still have some potential toxicity and lack specific targeting and disposition. This explains, in part, why their translation into the clinic has progressed only incrementally, which poses the need for more research to focus on addressing such translational limitations. This review summarizes the main properties of liposomes, their current status in cancer therapy, and their limitations and challenges to achieving maximal therapeutic efficacy.
Collapse
Affiliation(s)
- Melody D. Fulton
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA 99164, USA
| | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
78
|
Bashiri G, Padilla MS, Swingle KL, Shepherd SJ, Mitchell MJ, Wang K. Nanoparticle protein corona: from structure and function to therapeutic targeting. LAB ON A CHIP 2023; 23:1432-1466. [PMID: 36655824 PMCID: PMC10013352 DOI: 10.1039/d2lc00799a] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/29/2022] [Indexed: 05/31/2023]
Abstract
Nanoparticle (NP)-based therapeutics have ushered in a new era in translational medicine. However, despite the clinical success of NP technology, it is not well-understood how NPs fundamentally change in biological environments. When introduced into physiological fluids, NPs are coated by proteins, forming a protein corona (PC). The PC has the potential to endow NPs with a new identity and alter their bioactivity, stability, and destination. Additionally, the conformation of proteins is sensitive to their physical and chemical surroundings. Therefore, biological factors and protein-NP-interactions can induce changes in the conformation and orientation of proteins in vivo. Since the function of a protein is closely connected to its folded structure, slight differences in the surrounding environment as well as the surface characteristics of the NP materials may cause proteins to lose or gain a function. As a result, this can alter the downstream functionality of the NPs. This review introduces the main biological factors affecting the conformation of proteins associated with the PC. Then, four types of NPs with extensive utility in biomedical applications are described in greater detail, focusing on the conformation and orientation of adsorbed proteins. This is followed by a discussion on the instances in which the conformation of adsorbed proteins can be leveraged for therapeutic purposes, such as controlling protein conformation in assembled matrices in tissue, as well as controlling the PC conformation for modulating immune responses. The review concludes with a perspective on the remaining challenges and unexplored areas at the interface of PC and NP research.
Collapse
Affiliation(s)
- Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA.
| | - Marshall S Padilla
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah J Shepherd
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA.
| |
Collapse
|
79
|
Lee H. Differences in protein distribution, conformation, and dynamics in hard and soft coronas: dependence on protein and particle electrostatics. Phys Chem Chem Phys 2023; 25:7496-7507. [PMID: 36853334 DOI: 10.1039/d2cp05936c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
We perform all-atom molecular dynamics simulations of a 9 nm-thick protein layer, which consists of serum albumin (SA) or a mixture of SA and immunoglobulin gamma-1, formed on 10 nm-sized cationic, anionic, and neutral polystyrene particles. More than half of the proteins are densely concentrated within a distance of ∼3 nm from the particle surface, while fewer proteins are broadly distributed in the range of 3-9 nm from the particle. This compares favorably with the experimental observations of a hard corona as the first layer adjacent to the particle and a soft corona as a loose protein-network. The conformation and diffusivity of the proteins vary in different positions of the layer, and are to an extent dependent on the protein and particle electrostatics. These, combined with free energy calculations, show that the protein and particle charges do not significantly modify the strength of protein-particle binding but do influence the distribution of proteins in the layer. In particular, a free protein more strongly binds to the complex of a protein and particle than to either one, showing the synergistic effect of already adsorbed proteins and a particle. This helps explain the experimental observation regarding the formation of a denser protein layer and the stronger protein-protein interaction in the hard corona than the soft corona.
Collapse
Affiliation(s)
- Hwankyu Lee
- Department of Chemical Engineering, Dankook University, Yongin-si, 16890, South Korea.
| |
Collapse
|
80
|
Spleis H, Sandmeier M, Claus V, Bernkop-Schnürch A. Surface design of nanocarriers: Key to more efficient oral drug delivery systems. Adv Colloid Interface Sci 2023; 313:102848. [PMID: 36780780 DOI: 10.1016/j.cis.2023.102848] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
As nanocarriers (NCs) can improve the solubility of drugs, prevent their degradation by gastrointestinal (GI) enzymes and promote their transport across the mucus gel layer and absorption membrane, the oral bioavailability of these drugs can be substantially enhanced. All these properties of NCs including self-emulsifying drug delivery systems (SEDDS), solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), liposomes, polymeric nanoparticles, inorganic nanoparticles and polymeric micelles depend mainly on their surface chemistry. In particular, interaction with food, digestive enzymes, bile salts and electrolytes, diffusion behaviour across the mucus gel layer and fate on the absorption membrane are determined by their surface. Bioinert surfaces limiting interactions with gastrointestinal fluid and content as well as with mucus, adhesive surfaces providing an intimate contact with the GI mucosa and absorption enhancing surfaces can be designed. Furthermore, charge converting surfaces shifting their zeta potential from negative to positive directly at the absorption membrane and surfaces providing a targeted drug release are advantageous. In addition to these passive surfaces, even active surfaces cleaving mucus glycoproteins on their way through the mucus gel layer can be created. Within this review, we provide an overview on these different surfaces and discuss their impact on the performance of NCs in the GI tract.
Collapse
Affiliation(s)
- Helen Spleis
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Matthias Sandmeier
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Victor Claus
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria.
| |
Collapse
|
81
|
Känkänen V, Fernandes M, Liu Z, Seitsonen J, Hirvonen SP, Ruokolainen J, Pinto JF, Hirvonen J, Balasubramanian V, Santos HA. Microfluidic preparation and optimization of sorafenib-loaded poly(ethylene glycol-block-caprolactone) nanoparticles for cancer therapy applications. J Colloid Interface Sci 2023; 633:383-395. [PMID: 36462264 DOI: 10.1016/j.jcis.2022.11.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/09/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
The use of amphiphilic block copolymers to generate colloidal delivery systems for hydrophobic drugs has been the subject of extensive research, with several formulations reaching the clinical development stages. However, to generate particles of uniform size and morphology, with high encapsulation efficiency, yield and batch-to-batch reproducibility remains a challenge, and various microfluidic technologies have been explored to tackle these issues. Herein, we report the development and optimization of poly(ethylene glycol)-block-(ε-caprolactone) (PEG-b-PCL) nanoparticles for intravenous delivery of a model drug, sorafenib. We developed and optimized a glass capillary microfluidic nanoprecipitation process and studied systematically the effects of formulation and process parameters, including different purification techniques, on product quality and batch-to-batch variation. The optimized formulation delivered particles with a spherical morphology, small particle size (dH < 80 nm), uniform size distribution (PDI < 0.2), and high drug loading degree (16 %) at 54 % encapsulation efficiency. Furthermore, the stability and in vitro drug release were evaluated, showing that sorafenib was released from the NPs in a sustained manner over several days. Overall, the study demonstrates a microfluidic approach to produce sorafenib-loaded PEG-b-PCL NPs and provides important insight into the effects of nanoprecipitation parameters and downstream processing on product quality.
Collapse
Affiliation(s)
- Voitto Känkänen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; Drug Carrier and Depot Systems, Bayer Oy, FI-20210 Turku, Finland.
| | - Micaela Fernandes
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; iMed-ULisboa, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal; Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan, 1, 9713 AV Groningen, the Netherlands
| | - Zehua Liu
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jani Seitsonen
- Nanomicroscopy Center, Aalto University, Puumiehenkuja, 2, FI-02150 Espoo, Finland
| | - Sami-Pekka Hirvonen
- Department of Chemistry, Faculty of Science, University of Helsinki, P.O. Box 55, 00014 Helsinki, Finland
| | - Janne Ruokolainen
- Nanomicroscopy Center, Aalto University, Puumiehenkuja, 2, FI-02150 Espoo, Finland
| | - João F Pinto
- iMed-ULisboa, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | | | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan, 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
82
|
PEGylated and functionalized polylactide-based nanocapsules: An overview. Int J Pharm 2023; 636:122760. [PMID: 36858134 DOI: 10.1016/j.ijpharm.2023.122760] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/08/2023] [Accepted: 02/17/2023] [Indexed: 03/03/2023]
Abstract
Polymeric nanocapsules (NC) are versatile mixed vesicular nanocarriers, generally containing a lipid core with a polymeric wall. They have been first developed over four decades ago with outstanding applicability in the cosmetic and pharmaceutical fields. Biodegradable polyesters are frequently used in nanocapsule preparation and among them, polylactic acid (PLA) derivatives and copolymers, such as PLGA and amphiphilic block copolymers, are widely used and considered safe for different administration routes. PLA functionalization strategies have been developed to obtain more versatile polymers and to allow the conjugation with bioactive ligands for cell-targeted NC. This review intends to provide steps in the evolution of NC since its first report and the recent literature on PLA-based NC applications. PLA-based polymer synthesis and surface modifications are included, as well as the use of NC as a novel tool for combined treatment, diagnostics, and imaging in one delivery system. Furthermore, the use of NC to carry therapeutic and/or imaging agents for different diseases, mainly cancer, inflammation, and infections is presented and reviewed. Constraints that impair translation to the clinic are discussed to provide safe and reproducible PLA-based nanocapsules on the market. We reviewed the entire period in the literature where the term "nanocapsules" appears for the first time until the present day, selecting original scientific publications and the most relevant patent literature related to PLA-based NC. We presented to readers a historical overview of these Sui generis nanostructures.
Collapse
|
83
|
Jin T, Coley CW, Alexander-Katz A. Adsorption of Biomimetic Amphiphilic Heteropolymers onto Graphene and Its Derivatives. Macromolecules 2023. [DOI: 10.1021/acs.macromol.2c02413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Affiliation(s)
- Tianyi Jin
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Connor W. Coley
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Alfredo Alexander-Katz
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
84
|
Elmowafy M, Shalaby K, Elkomy MH, Alsaidan OA, Gomaa HAM, Abdelgawad MA, Mostafa EM. Polymeric Nanoparticles for Delivery of Natural Bioactive Agents: Recent Advances and Challenges. Polymers (Basel) 2023; 15:1123. [PMID: 36904364 PMCID: PMC10007077 DOI: 10.3390/polym15051123] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
In the last few decades, several natural bioactive agents have been widely utilized in the treatment and prevention of many diseases owing to their unique and versatile therapeutic effects, including antioxidant, anti-inflammatory, anticancer, and neuroprotective action. However, their poor aqueous solubility, poor bioavailability, low GIT stability, extensive metabolism as well as short duration of action are the most shortfalls hampering their biomedical/pharmaceutical applications. Different drug delivery platforms have developed in this regard, and a captivating tool of this has been the fabrication of nanocarriers. In particular, polymeric nanoparticles were reported to offer proficient delivery of various natural bioactive agents with good entrapment potential and stability, an efficiently controlled release, improved bioavailability, and fascinating therapeutic efficacy. In addition, surface decoration and polymer functionalization have opened the door to improving the characteristics of polymeric nanoparticles and alleviating the reported toxicity. Herein, a review of the state of knowledge on polymeric nanoparticles loaded with natural bioactive agents is presented. The review focuses on frequently used polymeric materials and their corresponding methods of fabrication, the needs of such systems for natural bioactive agents, polymeric nanoparticles loaded with natural bioactive agents in the literature, and the potential role of polymer functionalization, hybrid systems, and stimuli-responsive systems in overcoming most of the system drawbacks. This exploration may offer a thorough idea of viewing the polymeric nanoparticles as a potential candidate for the delivery of natural bioactive agents as well as the challenges and the combating tools used to overcome any hurdles.
Collapse
Affiliation(s)
- Mohammed Elmowafy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Khaled Shalaby
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Mohammed H. Elkomy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Hesham A. M. Gomaa
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
| |
Collapse
|
85
|
Jumai'an E, Zhang L, Bevan MA. Blood Protein Exclusion from Polymer Brushes. ACS NANO 2023; 17:2378-2386. [PMID: 36669160 DOI: 10.1021/acsnano.2c09332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
We report interactions between adsorbed copolymers of poly(ethylene glycol) (PEG) in the presence of two abundant blood proteins, serum albumin and an immunoglobulin G, up to physiological blood concentrations. We directly and nonintrusively measure interactions between PEG triblock copolymers (PEG-PPO-PEG) adsorbed to hydrophobic colloids and surfaces using Total Internal Reflection Microscopy, which provides kT- and nanometer-scale resolution of interaction potentials (energy vs separation). In the absence of protein, adsorbed PEG copolymer repulsion is consistent with dimensions and architectures of PEG brushes on both colloids and surfaces. In the presence of proteins, we observe concentration dependent depletion attraction and no change to brush repulsion, indicating protein exclusion from PEG brushes. Because positive and negative protein adsorption are mutually exclusive, our observations of concentration dependent depletion attraction with no change to brush repulsion unambiguously indicate the absence of protein coronas at physiological protein concentrations. These findings demonstrate a direct sensitive approach to determine interactions between proteins and particle/surface coatings important to diverse biotechnology applications.
Collapse
Affiliation(s)
- Eugenie Jumai'an
- Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland21218, United States
| | - Lechuan Zhang
- Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland21218, United States
| | - Michael A Bevan
- Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland21218, United States
| |
Collapse
|
86
|
Lebleu C, Plet L, Moussy F, Gitton G, Da Costa Moreira R, Guduff L, Burlot B, Godiveau R, Merry A, Lecommandoux S, Errasti G, Philippe C, Delacroix T, Chakrabarti R. Improving aqueous solubility of paclitaxel with polysarcosine-b-poly(γ-benzyl glutamate) nanoparticles. Int J Pharm 2023; 631:122501. [PMID: 36529355 DOI: 10.1016/j.ijpharm.2022.122501] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
New stealth amphiphilic copolymers based on polysarcosine (PSar) rather than poly(ethylene glycol) (PEG) have gained more attention for their use as excipients in nanomedicine. In this study, several polysarcosine-b-poly(γ-benzyl glutamate) (PSar-b-PGluOBn) block copolymers were synthesized by ring opening polymerization (ROP) of the respective N-carboxyanhydrides (NCAs) and were characterized by Fourier-transform infrared spectroscopy (FTIR), proton nuclear magnetic resonance (1H NMR) and size-exclusion chromatography (SEC). Copolymers had different PGluOBn block configuration (racemic L/D, pure L or pure D), degrees of polymerization of PSar between 28 and 76 and PGluOBn between 9 and 93, molar masses (Mn) between 5.0 and 24.6 kg.mol-1 and dispersities (Đ) lower than 1.4. Nanoparticles of PSar-b-PGluOBn loaded with paclitaxel (PTX), a hydrophobic anti-cancer drug, were obtained by nanoprecipitation. Their hydrodynamic diameter (Dh) ranged from 27 to 118 nm with polydispersity indexes (PDI) between 0.01 and 0.20, as determined by dynamic light scattering (DLS). Their morphology was more spherical for copolymers with a racemic L/D PGluOBn block configuration synthesized at 5 °C. PTX loading efficiency was between 63 and 92 % and loading contents between 7 and 15 %. Using PSar-b-PGluOBn copolymers as excipients, PTX apparent water-solubility was significantly improved by a factor up to 6600 to 660 µg.mL-1.
Collapse
Affiliation(s)
- Coralie Lebleu
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | - Laetitia Plet
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | - Florène Moussy
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | - Gaëtan Gitton
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | | | - Ludmilla Guduff
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | - Barbara Burlot
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | | | - Aïnhoa Merry
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | | | - Gauthier Errasti
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | | | - Thomas Delacroix
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France
| | - Raj Chakrabarti
- PMC Isochem SAS, 32, rue Lavoisier F-91710, Vert-Le-Petit, France; Chakrabarti Advanced Technology, LLC, PMC Group Building, 1288 Route 73, Ste 110, Mount Laurel, NJ 08054, USA.
| |
Collapse
|
87
|
Pontico M, Conte M, Petronella F, Frantellizzi V, De Feo MS, Di Luzio D, Pani R, De Vincentis G, De Sio L. 18F-fluorodeoxyglucose ( 18F-FDG) Functionalized Gold Nanoparticles (GNPs) for Plasmonic Photothermal Ablation of Cancer: A Review. Pharmaceutics 2023; 15:319. [PMID: 36839641 PMCID: PMC9967497 DOI: 10.3390/pharmaceutics15020319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
The meeting and merging between innovative nanotechnological systems, such as nanoparticles, and the persistent need to outperform diagnostic-therapeutic approaches to fighting cancer are revolutionizing the medical research scenario, leading us into the world of nanomedicine. Photothermal therapy (PTT) is a non-invasive thermo-ablative treatment in which cellular hyperthermia is generated through the interaction of near-infrared light with light-to-heat converter entities, such as gold nanoparticles (GNPs). GNPs have great potential to improve recovery time, cure complexity, and time spent on the treatment of specific types of cancer. The development of gold nanostructures for photothermal efficacy and target selectivity ensures effective and deep tissue-penetrating PTT with fewer worries about adverse effects from nonspecific distributions. Regardless of the thriving research recorded in the last decade regarding the multiple biomedical applications of nanoparticles and, in particular, their conjugation with drugs, few works have been completed regarding the possibility of combining GNPs with the cancer-targeted pharmaceutical fluorodeoxyglucose (FDG). This review aims to provide an actual scenario on the application of functionalized GNP-mediated PTT for cancer ablation purposes, regarding the opportunity given by the 18F-fluorodeoxyglucose (18F-FDG) functionalization.
Collapse
Affiliation(s)
- Mariano Pontico
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00185 Rome, Italy
| | - Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00185 Rome, Italy
| | - Francesca Petronella
- Institute of Crystallography CNR-IC, National Research Council of Italy, Monterotondo, 00015 Rome, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00185 Rome, Italy
| | - Maria Silvia De Feo
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00185 Rome, Italy
| | - Dario Di Luzio
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00185 Rome, Italy
| | - Roberto Pani
- Department of Medico-Surgical Sciences and Biotechnologies, Research Center for Biophotonics, Sapienza University of Rome, 04100 Latina, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00185 Rome, Italy
| | - Luciano De Sio
- Department of Medico-Surgical Sciences and Biotechnologies, Research Center for Biophotonics, Sapienza University of Rome, 04100 Latina, Italy
| |
Collapse
|
88
|
Toro-Mendoza J, Maio L, Gallego M, Otto F, Schulz F, Parak WJ, Sanchez-Cano C, Coluzza I. Bioinspired Polyethylene Glycol Coatings for Reduced Nanoparticle-Protein Interactions. ACS NANO 2023; 17:955-965. [PMID: 36602983 DOI: 10.1021/acsnano.2c05682] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Nanoparticles (NPs) and other engineered nanomaterials have great potential as nanodrugs or nanomedical devices for biomedical applications. However, the adsorption of proteins in blood circulation or similar physiological fluids can significantly alter the surface properties and therapeutic response induced by most nanomaterials. For example, interaction with proteins can change the bloodstream circulation time and availability of therapeutic NPs or hinder the accumulation in their desired target organs. Proteins can also trigger or prevent agglomeration. By combining experimental and computational approaches, we have developed NPs carrying polyethylene glycol (PEG) polymeric coatings that mimic the surface charge distribution of proteins typically found in blood, which are known to show low aggregation under normal blood conditions. Here, we show that NPs with coatings based on apoferritin or human serum albumin display better antifouling properties and weaker protein interaction compared to similar NPs carrying conventional PEG polymeric coatings.
Collapse
Affiliation(s)
- Jhoan Toro-Mendoza
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014Donostia-San Sebastián, Spain
| | - Lucia Maio
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014Donostia-San Sebastián, Spain
| | - Marta Gallego
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014Donostia-San Sebastián, Spain
| | - Ferdinand Otto
- Universität Hamburg, Luruper Chaussee 149, 22607Hamburg, Germany
| | - Florian Schulz
- Universität Hamburg, Luruper Chaussee 149, 22607Hamburg, Germany
| | - Wolfgang J Parak
- Universität Hamburg, Luruper Chaussee 149, 22607Hamburg, Germany
| | - Carlos Sanchez-Cano
- Ikerbasque, Basque Foundation for Science, Plaza de Euskadi 5, Bilbao48009, Spain
- Donostia International Physics Center (DIPC)Paseo Manuel de Lardizabal, 4, 20018Donostia/San Sebastian, Gipuzkoa, Spain
| | - Ivan Coluzza
- Ikerbasque, Basque Foundation for Science, Plaza de Euskadi 5, Bilbao48009, Spain
- BCMaterials, Bld. Martina Casiano, Third Floor, UPV/EHU Science Park, Barrio Sarriena s/n, 48940Leioa, Spain
| |
Collapse
|
89
|
Van Gheluwe L, David S, Buchy E, Chourpa I, Munnier E. Smart PEG-Block-PLA/PLA Nanosystems: Impact of the Characteristics of the Polymer Blend on the Redox Responsiveness. MATERIALS (BASEL, SWITZERLAND) 2023; 16:539. [PMID: 36676276 PMCID: PMC9864163 DOI: 10.3390/ma16020539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
Nanocarriers (NCs) were designed from three polymer blends (B1, B2 and B3) and investigated as smart drug delivery systems (SDDS). The blends are composed of a "smart" copolymer, where methoxy poly(ethylene glycol) and poly(lactic acid) are connected via a redox-responsive disulfide bond (mPEG-SS-PLA), and of a "conventional" polymer, poly(lactic acid) (PLA). They differ by mPEG-SS-PLA/PLA ratio and PLA molecular weight. Nanoprecipitation was used to prepare NCs. Three concentrations were tested, and fluorescent dye Nile red (NR) was used as a model payload. The results show that the characteristics of the NCs, such as size and drug release kinetics, are influenced by the type of blend and the concentration used during the nanoprecipitation process. The more redox-responsive blend was B2 (ratio 1:3, PLA 5 kDa) at 16 mg/mL: the quantity of NR released was tripled upon 24 h of incubation in a reducing medium. This study reveals that the amount of disulfide bonds present in a NC is not the only parameter to be considered to design an SDDS. The stability of the SDDS in a presumably non-stimulating environment is also important to limit uncontrolled release during storage or in the body before the biological target is reached.
Collapse
Affiliation(s)
- Louise Van Gheluwe
- EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Faculté de Pharmacie, 31 Avenue Monge, 37200 Tours, France
| | - Stephanie David
- EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Faculté de Pharmacie, 31 Avenue Monge, 37200 Tours, France
| | - Eric Buchy
- Laboratoires Eriger, 33 rue Augustin Fresnel, 37170 Chambray les Tours, France
| | - Igor Chourpa
- EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Faculté de Pharmacie, 31 Avenue Monge, 37200 Tours, France
| | - Emilie Munnier
- EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Faculté de Pharmacie, 31 Avenue Monge, 37200 Tours, France
| |
Collapse
|
90
|
Ghasemzadeh T, Hasannia M, Abnous K, Taghdisi SM, Nekooei S, Nekooei N, Ramezani M, Alibolandi M. Preparation of targeted theranostic red blood cell membranes-based nanobubbles for treatment of colon adenocarcinoma. Expert Opin Drug Deliv 2023; 20:131-143. [PMID: 36427011 DOI: 10.1080/17425247.2022.2152792] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Designing and fabrication of theranostic systems based on nanoscale gaseous vesicular systems, named nanobubbles (NBs), attracted enormous interest in recent years. Biomimetic vesicular platform (V-RBC-M) can improve the pharmacokinetics of the prepared platform due to augmented circulation half-life, desirable biodegradability and biocompatibility and reduced immunogenicity. METHODS V-RBC-M were used for the encapsulation of lipophilic camptothecin (CPT) in the bilayer of vesicles through top-down method, followed by filling the core of V-RBC-M with inert SF6 gas to fabricate NBs with ultrasonic contrast enhancement capability (SF6-NB-CPT). In the next step, targeted NBs were formed via decoration of MUC1 aptamer on the surface of NBs (Apt-SF6-NB-CPT). RESULTS The designed bio-NBs indicated high encapsulation efficiency and the sustained release of CPT at pH 7.4. In vitro study demonstrated higher cellular uptake and cytotoxicity of Apt-SF6-NB-CPT compared to SF6-NB-CPT in MUC1-overexpressing cells (C26). In vivo antitumor efficacy of the prepared NBs on C26 bearing BALB/c mice showed greater therapeutic efficacy and survival rate for Apt-SF6-NB-CPT. In this regard, SF6-NB-CPT showed 58% tumor growth suppression while Apt-SF6-NB-CPT system provided 95% tumor growth suppression. Furthermore, echogenic capability of SF6-NB-CPT was demonstrated through in vitro and in vivo ultrasonic imaging. CONCLUSIONS Our finding demonstrated that the prepared targeted NBs are a promising theranostic platform with effective therapeutic and diagnotic potentials.
Collapse
Affiliation(s)
- Tahoora Ghasemzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maliheh Hasannia
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
91
|
Alshareeda AT, Nur Khatijah MZ, Al-Sowayan BS. Nanotechnology: A revolutionary approach to prevent breast cancer recurrence. Asian J Surg 2023; 46:13-17. [PMID: 35361551 DOI: 10.1016/j.asjsur.2022.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/17/2021] [Accepted: 03/03/2022] [Indexed: 12/16/2022] Open
Abstract
Breast cancer is the most prevalent malignancy in women worldwide and one of the deadliest after lung cancer. Currently, standard treatment approaches for breast cancers are surgery accompanied by chemotherapy or radiotherapy. Cancer local recurrence after mastectomy is commonly considered as being a poor prognostic predictor. There have been advancements in the procedures utilized for breast reconstruction following mastectomy, much as there have been advancements in the early diagnosis and treatment of breast cancer. For the last decade, developing nanotechnology applications for cancer therapies has had much focus. The benefits granted by nanotechnologies via enhancing biological processes and promoting better biomaterial compatibility, as well as generating functionalized tissues, transpire exciting possibilities. Modified nanomedicine may introduce tremendous improvements to the fields of breast cancer recurrence through implants. It can modify the surfaces of implants to optimize tissue growth, thus minimizing inflammation and unsatisfactory results. Here we discuss new nanotechnology advancements and incorporate them into breast reconstruction surgeries following mastectomy or lumpectomy. In addtion, we repurpose old technologies, like growth factor therapies using nanotechnology for more efficient delivery.
Collapse
Affiliation(s)
- Alaa T Alshareeda
- The Saudi Biobank Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; Stem Cells and Regenerative Medicine Unit, Cellular Therapy and Cancer Research Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.
| | - M Z Nur Khatijah
- Department of Cell Physiology, Jikei University School of Medicine, Tokyo, Japan.
| | - Batla S Al-Sowayan
- Stem Cells and Regenerative Medicine Unit, Cellular Therapy and Cancer Research Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.
| |
Collapse
|
92
|
Pershina AG, Demin AM, Perekucha NA, Brikunova OY, Efimova LV, Nevskaya KV, Vakhrushev AV, Zgoda VG, Uimin MA, Minin AS, Malkeyeva D, Kiseleva E, Zima AP, Krasnov VP, Ogorodova LM. Peptide ligands on the PEGylated nanoparticle surface and human serum composition are key factors for the interaction between immune cells and nanoparticles. Colloids Surf B Biointerfaces 2023; 221:112981. [DOI: 10.1016/j.colsurfb.2022.112981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
|
93
|
Xie Y, Zhong Z, Wang W, Huang Y, Wu C, Pan X, Huang Z. Three Different Interaction Patterns between MCM-41 and Proteins. Int J Mol Sci 2022; 23:15850. [PMID: 36555490 PMCID: PMC9781137 DOI: 10.3390/ijms232415850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
As one of the most studied mesoporous silica nanoparticles (MSNs) in drug delivery systems, Mobil Composition of Matter No. 41 (MCM-41) possesses unique properties including perfect channel architecture, excellent load capacity, and good biocompatibility. However, the applications of MCM-41 nanoparticles in drug delivery have not yet been industrialized, due to the interaction between MCM-41 and biomolecules (especially proteins) that affect their in vivo behaviors after dosing. To investigate the interactions between MCM-41 and proteins, this study selected bovine serum albumin (BSA), lysozyme (Lyso), and bovine hemoglobin (BHb) as model proteins and characterized the ultraviolet-visible, fluorescence, circular dichroism spectra and the protein adsorption of MCM-41-protein complex. The UV-Vis spectra exhibited the different absorption increment degrees of three proteins. The fluorescence spectra showed that the fluorescence intensity of proteins changed by different trends. The CD spectra indicated that the secondary structure changes were ranked as BSA > Lyso > BHb, which is consistent with the protein’s adsorption capability on MCM-41. It was shown that there were three different patterns of MCM-41-proteins interactions. The hydrophilic and low-charged BSA followed the strong interaction pattern, the hydrophilic but heavily charged Lyso followed the moderate interaction pattern, and the hydrophobic BHb followed the weak interaction pattern. Different interaction patterns would lead to different effects on the structural properties of proteins, the surface chemistry of MCM-41, and the absorption capability of proteins on MCM-41. We believe our study will provide a better insight into the application of MCM-41 nanoparticles in drug delivery systems.
Collapse
Affiliation(s)
- Yuke Xie
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Ziqiao Zhong
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Ying Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| |
Collapse
|
94
|
El-Baz N, Nunn BM, Bates PJ, O’Toole MG. The Impact of PEGylation on Cellular Uptake and In Vivo Biodistribution of Gold Nanoparticle MRI Contrast Agents. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120766. [PMID: 36550972 PMCID: PMC9774698 DOI: 10.3390/bioengineering9120766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Gold nanoparticles (GNPs) have immense potential in biomedicine, but understanding their interactions with serum proteins is crucial as it could change their biological profile due to the formation of a protein corona, which could then affect their ultimate biodistribution in the body. Grafting GNPs with polyethylene glycol (PEG) is a widely used practice in research in order to decrease opsonization of the particles by serum proteins and to decrease particle uptake by the mononuclear phagocyte system. We investigated the impact of PEGylation on the formation of protein coronae and the subsequent uptake by macrophages and MDA-MB-231 cancer cells. Furthermore, we investigated the in vivo biodistribution in xenograft tumor-bearing mice using a library of 4 and 10 nm GNPs conjugated with a gadolinium chelate as MRI contrast agent, cancer-targeting aptamer AS1411 (or CRO control oligonucleotide), and with or without PEG molecules of different molecular weight (Mw: 1, 2, and 5 kDa). In vitro results showed that PEG failed to decrease the adsorption of proteins; moreover, the cellular uptake by macrophage cells was contingent on the different configurations of the aptamers and the length of the PEG chain. In vivo biodistribution studies showed that PEG increased the uptake by tumor cells for some GNPs, albeit it did not decrease the uptake of GNPs by macrophage-rich organs.
Collapse
Affiliation(s)
- Nagwa El-Baz
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40292, USA
| | - Betty M. Nunn
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Paula J. Bates
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Martin G. O’Toole
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
- Correspondence:
| |
Collapse
|
95
|
Chien Y, Hsiao YJ, Chou SJ, Lin TY, Yarmishyn AA, Lai WY, Lee MS, Lin YY, Lin TW, Hwang DK, Lin TC, Chiou SH, Chen SJ, Yang YP. Nanoparticles-mediated CRISPR-Cas9 gene therapy in inherited retinal diseases: applications, challenges, and emerging opportunities. J Nanobiotechnology 2022; 20:511. [DOI: 10.1186/s12951-022-01717-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/23/2022] [Indexed: 12/04/2022] Open
Abstract
AbstractInherited Retinal Diseases (IRDs) are considered one of the leading causes of blindness worldwide. However, the majority of them still lack a safe and effective treatment due to their complexity and genetic heterogeneity. Recently, gene therapy is gaining importance as an efficient strategy to address IRDs which were previously considered incurable. The development of the clustered regularly-interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) system has strongly empowered the field of gene therapy. However, successful gene modifications rely on the efficient delivery of CRISPR-Cas9 components into the complex three-dimensional (3D) architecture of the human retinal tissue. Intriguing findings in the field of nanoparticles (NPs) meet all the criteria required for CRISPR-Cas9 delivery and have made a great contribution toward its therapeutic applications. In addition, exploiting induced pluripotent stem cell (iPSC) technology and in vitro 3D retinal organoids paved the way for prospective clinical trials of the CRISPR-Cas9 system in treating IRDs. This review highlights important advances in NP-based gene therapy, the CRISPR-Cas9 system, and iPSC-derived retinal organoids with a focus on IRDs. Collectively, these studies establish a multidisciplinary approach by integrating nanomedicine and stem cell technologies and demonstrate the utility of retina organoids in developing effective therapies for IRDs.
Collapse
|
96
|
Nierenberg D, Flores O, Fox D, Sip YYL, Finn CM, Ghozlan H, Cox A, Coathup M, McKinstry KK, Zhai L, Khaled AR. Macromolecules Absorbed from Influenza Infection-Based Sera Modulate the Cellular Uptake of Polymeric Nanoparticles. Biomimetics (Basel) 2022; 7:biomimetics7040219. [PMID: 36546919 PMCID: PMC9775140 DOI: 10.3390/biomimetics7040219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
Optimizing the biological identity of nanoparticles (NPs) for efficient tumor uptake remains challenging. The controlled formation of a protein corona on NPs through protein absorption from biofluids could favor a biological identity that enables tumor accumulation. To increase the diversity of proteins absorbed by NPs, sera derived from Influenza A virus (IAV)-infected mice were used to pre-coat NPs formed using a hyperbranched polyester polymer (HBPE-NPs). HBPE-NPs, encapsulating a tracking dye or cancer drug, were treated with sera from days 3-6 of IAV infection (VS3-6), and uptake of HBPE-NPs by breast cancer cells was examined. Cancer cells demonstrated better uptake of HBPE-NPs pre-treated with VS3-6 over polyethylene glycol (PEG)-HBPE-NPs, a standard NP surface modification. The uptake of VS5 pre-treated HBPE-NPs by monocytic cells (THP-1) was decreased over PEG-HBPE-NPs. VS5-treated HBPE-NPs delivered a cancer drug more efficiently and displayed better in vivo distribution over controls, remaining stable even after interacting with endothelial cells. Using a proteomics approach, proteins absorbed from sera-treated HBPE-NPs were identified, such as thrombospondin-1 (TSP-1), that could bind multiple cancer cell receptors. Our findings indicate that serum collected during an immune response to infection is a rich source of macromolecules that are absorbed by NPs and modulate their biological identity, achieving rationally designed uptake by targeted cell types.
Collapse
Affiliation(s)
- Daniel Nierenberg
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Orielyz Flores
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - David Fox
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
- Department of Chemistry, College of Science, University of Central Florida, Orlando, FL 32816, USA
| | - Yuen Yee Li Sip
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
- Department of Materials Science and Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, FL 32816, USA
| | - Caroline M. Finn
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Heba Ghozlan
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Amanda Cox
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Melanie Coathup
- Biionix Cluster and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Karl Kai McKinstry
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
| | - Lei Zhai
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
- Department of Chemistry, College of Science, University of Central Florida, Orlando, FL 32816, USA
- Department of Materials Science and Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, FL 32816, USA
| | - Annette R. Khaled
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- NanoScience Technology Science Center, University of Central Florida, Orlando, FL 32826, USA
- Correspondence: ; Tel.: +1-407-266-7035
| |
Collapse
|
97
|
Fahmi A, Abdur-Rahman M, Mahareek O, shemis MA. Synthesis, characterization, and cytotoxicity of doxorubicin-loaded polycaprolactone nanocapsules as controlled anti-hepatocellular carcinoma drug release system. BMC Chem 2022; 16:95. [DOI: 10.1186/s13065-022-00888-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/29/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Free doxorubicin (Dox) is used as a chemotherapeutic agent against hepatocellular carcinoma (HCC), but it results in cardiotoxicty as a major side effect. Hence, a controlled Dox drug delivery system is extremely demanded.
Methods
Dox was loaded into the non-toxic biodegradable polycaprolactone (PCL) nanocapsules using the double emulsion method. Characterization of Dox-PCL nanocapsules was done using transmission electron microscopy and dynamic light scattering. Encapsulation efficiency and drug loading capacity were quantified using UV–visible spectrophotometry. Drug release was investigated in vitro at both normal (7.4) and cancer (4.8) pHs. Cytotoxicity of Dox-PCL nanocapsules against free Dox was evaluated using the MTT test on normal (Vero) and hepatic cancer (HepG2) cell lines.
Results
Spherical nanocapsules (212 ± 2 nm) were succeffully prepared with a zeta potential of (-22.3 ± 2 mv) and a polydisperse index of (0.019 ± 0.01) with a narrow size distribution pattern. The encapsulation efficiency was (73.15 ± 4%) with a drug loading capacity of (16.88 ± 2%). Importantlly, Dox-release from nanocapsules was faster at cancer pH (98%) than at physiological pH (26%). Moreover, although Dox-PCL nanocapsules were less toxic on the normal cell line (GI 50 = 17.99 ± 8.62 µg/ml) than free Dox (GI 50 = 16.53 ± 1.06 µg/ml), the encapsulated Dox showed higher toxic effect on cancer HepG2 cells compared to that caused by the free drug (GI 50 = 2.46 ± 0.49 and 4.22 ± 0.04 µg/ml, respectively).
Conclusion
The constructed Dox-PCL nanocapsules constitute a potentially controlled anti-HCC therapy with minimal systemic exposure.
Graphical Abstract
Collapse
|
98
|
Bueno-Alejo C, Santana Vega M, Chaplin AK, Farrow C, Axer A, Burley GA, Dominguez C, Kara H, Paschalis V, Tubasum S, Eperon IC, Clark AW, Hudson AJ. Surface Passivation with a Perfluoroalkane Brush Improves the Precision of Single-Molecule Measurements. ACS APPLIED MATERIALS & INTERFACES 2022; 14:49604-49616. [PMID: 36306432 PMCID: PMC9650645 DOI: 10.1021/acsami.2c16647] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/17/2022] [Indexed: 06/16/2023]
Abstract
Single-molecule imaging is invaluable for investigating the heterogeneous behavior and interactions of biological molecules. However, an impediment to precise sampling of single molecules is the irreversible adsorption of components onto the surfaces of cover glasses. This causes continuous changes in the concentrations of different molecules dissolved or suspended in the aqueous phase from the moment a sample is dispensed, which will shift, over time, the position of chemical equilibria between monomeric and multimeric components. Interferometric scattering microscopy (iSCAT) is a technique in the single-molecule toolkit that has the capability to detect unlabeled proteins and protein complexes both as they adsorb onto and desorb from a glass surface. Here, we examine the reversible and irreversible interactions between a number of different proteins and glass via analysis of the adsorption and desorption of protein at the single-molecule level. Furthermore, we present a method for surface passivation that virtually eliminates irreversible adsorption while still ensuring the residence time of molecules on surfaces is sufficient for detection of adsorption by iSCAT. By grafting high-density perfluoroalkane brushes on cover-glass surfaces, we observe approximately equal numbers of adsorption and desorption events for proteins at the measurement surface (±1%). The fluorous-aqueous interface also prevents the kinetic trapping of protein complexes and assists in establishing a thermodynamic equilibrium between monomeric and multimeric components. This surface passivation approach is valuable for in vitro single-molecule experiments using iSCAT microscopy because it allows for continuous monitoring of adsorption and desorption of protein without either a decline in detection events or a change in sample composition due to the irreversible binding of protein to surfaces.
Collapse
Affiliation(s)
- Carlos
J. Bueno-Alejo
- School
of Chemistry, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Marina Santana Vega
- School
of Engineering, Advanced Research Centre, University of Glasgow, 11 Chapel Lane, Glasgow G11 6EW, United Kingdom
| | - Amanda K. Chaplin
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
- Department
of Molecular and Cellular Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Chloe Farrow
- School
of Chemistry, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Alexander Axer
- Strathclyde
Centre for Molecular Bioscience & Department of Pure & Applied
Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| | - Glenn A. Burley
- Strathclyde
Centre for Molecular Bioscience & Department of Pure & Applied
Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| | - Cyril Dominguez
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
- Department
of Molecular and Cellular Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Hesna Kara
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
- Department
of Molecular and Cellular Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Vasileios Paschalis
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
- Department
of Molecular and Cellular Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Sumera Tubasum
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
- Department
of Molecular and Cellular Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Ian C. Eperon
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
- Department
of Molecular and Cellular Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| | - Alasdair W. Clark
- School
of Engineering, Advanced Research Centre, University of Glasgow, 11 Chapel Lane, Glasgow G11 6EW, United Kingdom
| | - Andrew J. Hudson
- School
of Chemistry, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
- Leicester
Institute of Structural & Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7HB, United Kingdom
| |
Collapse
|
99
|
Zhong Z, Chen Z, Xie Y, Wang W, Huang Z, Huang Y, Wu C, Pan X. The Effect of Sulfobetaine Coating in Inhibiting the Interaction between Lyotropic Liquid Crystalline Nanogels and Proteins. Gels 2022; 8:653. [PMID: 36286154 PMCID: PMC9602168 DOI: 10.3390/gels8100653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 11/04/2022] Open
Abstract
The injective lyotropic liquid crystalline nanogels (LLCNs) were widely used in drug delivery systems. But when administered in vivo, LLCNs exposed to the biological environment interact with proteins. Recently, it has been shown that nanoparticles coated with zwitterions can inhibit their interaction with proteins. Thus, in this study, the interaction between proteins and LLCNs coated with the zwitterionic material sulfobetaine (GLLCNs@HDSB) was investigated using bovine serum albumin (BSA) as a model protein. Interestingly, it was found that GLLCNs@HDSB at higher concentrations (≥0.8 mg/mL) could block its interaction with BSA, but not at lower concentrations (<0.8 mg/mL), according to the results of ultraviolet, fluorescence, and circular dichroism spectra. In the ultraviolet spectra, the absorbance of GLLCNs@HDSB (0.8 mg/mL) was 1.9 times higher than that without the sulfobetaine coating (GLLCNs) after incubation with protein; the fluorescence quenching intensity of GLLCNs@HDSB was conversely larger than that of the GLLCNs; in circular dichroism spectra, the ellipticity value of GLLCNs@HDSB was significantly smaller than that of the GLLCNs, and the change in GLLCNs@HDSB was 10 times higher than that of the GLLCNs. Generally, nanoparticles coated with sulfobetaine can inhibit their interaction with proteins, but in this study, LLCNs showed a concentration-dependent inhibitory effect. It could be inferred that in contrast to the surface of nanoparticles covered with sulfobetaine in other cases, the sulfobetaine in this study interacted with the LLCNs and was partially inserted into the hydrophobic region of the LLCNs. In conclusion, this study suggests that coating-modified nanoparticles do not necessarily avoid interacting with proteins, and we should also study coating-modified nanoparticles interacting with proteins both in vitro and in vivo. In the future, finding a coating material to completely inhibit the interaction between LLCNs and proteins will generate a great impetus to promote the clinical transformation of LLCNs.
Collapse
Affiliation(s)
- Ziqiao Zhong
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Zhiwei Chen
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Yuke Xie
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Ying Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
100
|
Zalba S, Ten Hagen TLM, Burgui C, Garrido MJ. Stealth nanoparticles in oncology: Facing the PEG dilemma. J Control Release 2022; 351:22-36. [PMID: 36087801 DOI: 10.1016/j.jconrel.2022.09.002] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022]
Abstract
Nanoparticles (Nps) have revolutionized the landscape of many treatments, by modifying not only pharmacokinetic properties of the encapsulated agent, but also providing a significant protection of the drug from non-desired interactions, and reducing side-effects of the enclosed therapeutic, enabling co-encapsulation of possibly synergistic compounds or activities, allowing a controlled release of content and improving the therapeutic effect. Nevertheless, in systemic circulation, Nps suffer a rapid removal by opsonisation and the action of Mononuclear phagocyte system (MPS). To overcome this problem, different polymers, in particular Polyethyleneglycol (PEG), have been used to cover the surface of these nanocarriers forming a hydrophilic layer that allows the delay of the removal. These advantages contrast with some drawbacks such as the difficulty to interact with cell membranes and the development of immunological reactions, conforming the known, "PEG dilemma". To address and minimize this phenomenon, different strategies have been applied. Therefore, this review aims to summarize the state of the art of Pegylation strategies, comment in depth on the principal characteristics of PEG and describe the main alternatives, which are the use of cleavable PEG, addition of different polymers or even use other derivatives of cell membranes to camouflage Nps.
Collapse
Affiliation(s)
- Sara Zalba
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy & Nutrition, University of Navarra; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology, and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Carmen Burgui
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy & Nutrition, University of Navarra
| | - María J Garrido
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy & Nutrition, University of Navarra; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|