1
|
Traeger A, Leiske MN. The Whole Is Greater than the Sum of Its Parts - Challenges and Perspectives in Polyelectrolytes. Biomacromolecules 2025; 26:5-32. [PMID: 39661745 PMCID: PMC11733940 DOI: 10.1021/acs.biomac.4c01061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
Polyelectrolytes offer unique properties for biological applications due to their charged nature and high water solubility. Here, the challenges in their synthesis and characterization techniques are reviewed, emphasizing that their strong interactions with the surrounding media and counterions must be considered when working with this interesting class of materials. Their potential in complexation for gene delivery, their unique stealth and anti-fouling properties, and their more specific interactions with amino acid transporters for cancer therapy are highlighted. The underlying mechanisms responsible for their biological efficacy, including the proton sponge effect for endosomal release and their interactions with cellular membranes, are addressed. For polyelectrolytes with a high level of usage, an overview is given of their historical context. This Perspective outlines the potential of polyelectrolytes for innovative applications in the field of biomedicine. Considering the physicochemical characteristics of this class of materials, this work strives to elucidate the distinctive properties and applications of polyelectrolytes.
Collapse
Affiliation(s)
- Anja Traeger
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, 07743 Jena, Germany
- Jena Center
for Soft Matter (JCSM), Friedrich Schiller
University Jena, 07743 Jena, Germany
| | - Meike N. Leiske
- Macromolecular
Chemistry, University of Bayreuth, 95447 Bayreuth, Germany
- Bavarian
Polymer Institute, 95447 Bayreuth, Germany
| |
Collapse
|
2
|
Li P, Li J, Cheng J, Huang J, Li J, Xiao J, Duan X. Hypoxia-responsive liposome enhances intracellular delivery of photosensitizer for effective photodynamic therapy. J Control Release 2025; 377:277-287. [PMID: 39561946 DOI: 10.1016/j.jconrel.2024.11.032] [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: 06/25/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Liposomes, especially polyethylene glycol (PEG)-modified long-circulating liposomes, have been approved for market use, due to good biocompatibility, passive tumor targeting, and sustained drug release. PEG-modified long-circulating liposomes address issues such as poor stability and rapid clearance by the reticuloendothelial system. However, they still face challenges like hindering drug uptake by tumor cells and preventing tumor penetration. Inspired by the hypoxic tumor microenvironment, we constructed a hypoxia-responsive liposome (PAO-L) to enhance the intracellular uptake and photodynamic therapy (PDT) effect of chlorin e6 (Ce6). The intelligent hypoxia-cleavable PEG-AZO-OA (PAO) was prepared by coupling PEG and octadecylamine (OA) to hypoxia-sensitive azobenzene-4,4'-dicarboxylic acid (AZO) through amide reaction. The synthesized PAO was further incorporated into Ce6-loaded liposomes to enhance the circulation stability, while promote the tumor penetration and internalization by the responsive shedding of PEG from liposome surface upon reaching the hypoxic tumor tissue. PAO-L mediated PDT significantly inhibited the growth of B16F10 and 4T1 tumors, as well as lung metastasis of 4T1 breast cancer. The excellent therapeutic effect and good tolerability make PAO-L a promising candidate for enhanced PDT.
Collapse
Affiliation(s)
- Peishan Li
- Department of General Surgery, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiaxin Li
- Department of General Surgery, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jinmei Cheng
- Department of General Surgery, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Junyi Huang
- Department of General Surgery, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; Department of Cardiology, Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jinhui Li
- Experimental Education/Administration Center, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Jisheng Xiao
- Department of General Surgery, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China; Department of Cardiology, Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Xiaopin Duan
- Department of General Surgery, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
3
|
Anand P, Zhang Y, Patil S, Kaur K. Metabolic Stability and Targeted Delivery of Oligonucleotides: Advancing RNA Therapeutics Beyond The Liver. J Med Chem 2025. [PMID: 39772535 DOI: 10.1021/acs.jmedchem.4c02528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Oligonucleotides have emerged as a formidable new class of nucleic acid therapeutics. Fully modified oligonucleotides exhibit enhanced metabolic stability and display successful clinical applicability for targets formerly considered "undruggable". Accumulating studies show that conjugation to targeting modalities of stabilized oligonucleotides, especially small interfering RNAs (siRNAs), has enabled robust delivery to intended cells/tissues. However, the major challenge in the field has been the stability and targeted delivery of oligonucleotides (siRNAs and antisense oligonucleotides (ASOs)) to extrahepatic tissues. In this Perspective, we review chemistry innovations and emerging delivery approaches that have revolutionized oligonucleotide drug discovery and development. We explore findings from both academia and industry that highlight the potential of oligonucleotides for indications involving different extrahepatic organs─including skeletal muscles, brain, lungs, skin, heart, adipose tissue, and eyes. In all, continued advances in chemistry coupled with conjugation-based approaches or novel administration routes will further advance the delivery of oligonucleotides to extrahepatic tissues.
Collapse
Affiliation(s)
- Puneet Anand
- Regeneron Genetic Medicines, Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Yu Zhang
- Regeneron Genetic Medicines, Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Spoorthi Patil
- Regeneron Genetic Medicines, Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Keerat Kaur
- Regeneron Genetic Medicines, Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| |
Collapse
|
4
|
Huang K, Liu Y, Miao H, Li Y, Fan Q, Zhang H, Zuo C, Zhu J, Zheng Q, Deng C, Sun Z, Tong Z. Nebulized Lipid Nanoparticles Based on Degradable Ionizable Glycerolipid for Potent Pulmonary mRNA Delivery. ACS NANO 2025. [PMID: 39754306 DOI: 10.1021/acsnano.4c13053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
As an advanced nucleic acid therapeutical modality, mRNA can express any type of protein in principle and thus holds great potential to prevent and treat various diseases. Despite the success in COVID-19 mRNA vaccines, direct local delivery of mRNA into the lung by inhalation would greatly reinforce the treatment of pulmonary pathogens and diseases. Herein, we developed lipid nanoparticles (LNPs) from degradable ionizable glycerolipids for potent pulmonary mRNA delivery via nebulization. A panel of proprietary ionizable glycerolipids with branched tails and five ester bonds were developed through a three-step esterification reaction, and LNPs formed from a lead glycerolipid identified as TG4C enabled highly efficient in vivo mRNA delivery via systemic administration with around 6-fold higher luciferase protein expression than commercial LNPs from SM102 and Dlin-MC3-DMA (MC3). Formulation screening revealed that LNPs formed at a TG4C:DOPE:cholesterol:DMG-PEG molar ratio of 50:10:38.5:1.5 (TG4C-LNPs4) had high stability against nebulization with slight changes of size distribution and mRNA encapsulation efficiency, and the nebulized TG4C-LNPs4 afforded an equivalent percentage of positive cells and a slightly lower EGFP fluorescence intensity in lung cell lines (A549, BEAS-2B). Following pulmonary delivery in mice, TG4C-LNPs4 induced efficient transfection in the majority of epithelial cells in the lung, leading to apparent bioluminescence evenly distributed in all five lung lobes. In an elastase-induced emphysema model in mice, TG4C-LNPs4 loaded with mRNA encoding hepatocyte growth factor could significantly suppress the secretion of inflammatory cytokines (IL-1β, IL-6, and TNF-α) in bronchoalveolar lavage fluid and counteract the alveoli wall thinning. Notably, partial substitution (25%) of cholesterol with budesonide, an anti-inflammatory glucocorticoid, in TG4C-LNPs4 generated equivalent protein expression and significantly improved therapeutic efficacy. Taken together, our study provides robust and high-performing nanovehicles based on degradable ionizable glycerolipids, enabling potently local mRNA delivery to the lung for the treatment of pulmonary diseases.
Collapse
Affiliation(s)
- Ke Huang
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Yuping Liu
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Hao Miao
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Yingwen Li
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Qianyi Fan
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Huanyu Zhang
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Chijian Zuo
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Jiafeng Zhu
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Qijun Zheng
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Materials, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Zhenhua Sun
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou 215125, China
| | - Zhenbo Tong
- Southeast University-Monash University Joint Research Institute, Suzhou 215125, China
| |
Collapse
|
5
|
Serrano A, Casares N, Trocóniz IF, Lozano T, Lasarte JJ, Zalba S, Garrido MJ. Foxp3 inhibitory peptide encapsulated in a novel CD25-targeted nanoliposome promotes efficient tumor regression in mice. Acta Pharmacol Sin 2025; 46:171-183. [PMID: 39075226 PMCID: PMC11695603 DOI: 10.1038/s41401-024-01338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/06/2024] [Indexed: 07/31/2024] Open
Abstract
P60, a Foxp3 inhibitory peptide, can hinder the regulatory T cell (Treg) activity and impair tumor proliferation. However, low systemic stability and poor specificity have led to daily dosing to achieve therapeutic effect. Therefore, this study aims to improve P60 stability and specific delivery through its encapsulation in liposomes targeting CD25, constitutively expressed in Tregs. P60 liposomes formulated with DSPE-PEG750 or DSPE-PEG2000 were incubated with DSPE-PEG2000-Maleimide micelles conjugated to Fab' fragments of anti-CD25 to develop two targeted formulations or immunoliposomes (IL): IL-P602000 (DSPE-PEG2000 only) and IL-P60750 (combining DSPE-PEG750 and DSPE-PEG2000). P60 encapsulation efficiency was 50%-60% irrespective of PEG chain length. Treg uptake was 2.5 and 14 times higher for IL-PEG750 compared with IL-PEG2000 and non-targeted liposomes, respectively, in in-vitro assays. In fact, IL-P60750 allowed CD8+ T cells ex-vivo proliferation in presence of Treg at doses 10-20 times lower than for free P60. Antitumor response of P60 and IL-P60750 in monotherapy and combined with anti-PD-1 was evaluated in MC38 and LLCOVA tumor bearing mice. In MC38 model, IL-P60750 monotherapy induced total tumor regression in 40% of mice reaching 100% for anti-PD-1 combination. This effect was associated with a significant increase in activated CD8+ T cells in tumors. Notably, IL-P60750 also inhibited human Treg in ex-vivo assay, showing the translational capability of this formulation. In conclusion, IL-P60750 formulated with different PEG chain lengths, has demonstrated antitumor efficacy by selective inhibition of Treg activity and enhances the effect of anti-PD1. Altogether, this novel IL represents a promising nanoplatform for cancer immunotherapies.
Collapse
Affiliation(s)
- Alejandro Serrano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
| | - Noelia Casares
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, CIMA, Pamplona, Spain
| | - Iñaki F Trocóniz
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
| | - Teresa Lozano
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, CIMA, Pamplona, Spain
| | - Juan J Lasarte
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, CIMA, Pamplona, Spain
| | - Sara Zalba
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain.
| | - María J Garrido
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain.
| |
Collapse
|
6
|
Cassani M, Fernandes S, Pagliari S, Cavalieri F, Caruso F, Forte G. Unraveling the Role of the Tumor Extracellular Matrix to Inform Nanoparticle Design for Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409898. [PMID: 39629891 PMCID: PMC11727388 DOI: 10.1002/advs.202409898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Indexed: 01/14/2025]
Abstract
The extracellular matrix (ECM)-and its mechanobiology-regulates key cellular functions that drive tumor growth and development. Accordingly, mechanotherapy is emerging as an effective approach to treat fibrotic diseases such as cancer. Through restoring the ECM to healthy-like conditions, this treatment aims to improve tissue perfusion, facilitating the delivery of chemotherapies. In particular, the manipulation of ECM is gaining interest as a valuable strategy for developing innovative treatments based on nanoparticles (NPs). However, further progress is required; for instance, it is known that the presence of a dense ECM, which hampers the penetration of NPs, primarily impacts the efficacy of nanomedicines. Furthermore, most 2D in vitro studies fail to recapitulate the physiological deposition of matrix components. To address these issues, a comprehensive understanding of the interactions between the ECM and NPs is needed. This review focuses on the main features of the ECM and its complex interplay with NPs. Recent advances in mechanotherapy are discussed and insights are offered into how its combination with nanomedicine can help improve nanomaterials design and advance their clinical translation.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Soraia Fernandes
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Stefania Pagliari
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Giancarlo Forte
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|
7
|
Lahouty M, Fadaee M, Shanehbandi D, Kazemi T. Exosome-driven nano-immunotherapy: revolutionizing colorectal cancer treatment. Mol Biol Rep 2024; 52:83. [PMID: 39724304 DOI: 10.1007/s11033-024-10157-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Colorectal cancer (CRC) ranks as the third most common cancer worldwide and remains a major cause of cancer-related deaths, necessitating the development of innovative therapeutic approaches beyond conventional treatment modalities. Conventional therapies, such as radiation, chemotherapy, and surgery, are hindered by challenges like imprecise targeting, substantial toxicity, and the development of resistance. Exosome-driven nano-immunotherapy has emerged as a groundbreaking approach that leverages the natural properties of exosomes-cell-derived vesicles known for their role in intercellular communication-to deliver therapeutic agents with high precision and specificity. This approach utilizes the natural ability of exosomes to serve as natural nanocarriers for various biomolecules, such as proteins, nucleic acids, and lipids, enabling precise drug delivery and immune modulation. Exosomes offer distinct advantages compared to traditional drug delivery systems, including their biocompatibility, capability to traverse biological barriers, and suitability for personalized medicine approaches. We evaluate the effectiveness of exosome-based therapies in comparison to traditional approaches, emphasizing their ability to achieve precise delivery, minimize systemic toxicity, and enhance treatment results. Despite their promise, several challenges remain, including the standardization of exosome isolation and production, optimization of cargo loading techniques, and ensuring safety and efficacy in clinical applications. By overcoming these obstacles and leveraging the distinctive characteristics of exosomes, exosome-driven nano-immunotherapy presents a promising avenue for more efficient therapeutic interventions.
Collapse
Affiliation(s)
- Masoud Lahouty
- Department of Microbiology and Virology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Manouchehr Fadaee
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
8
|
Koehler JK, Schmager S, Schnur J, Gedda L, Edwards K, Heerklotz H, Massing U. Novel thermosensitive small multilamellar lipid nanoparticles with promising release characteristics made by dual centrifugation. Eur J Pharm Sci 2024; 206:106999. [PMID: 39730031 DOI: 10.1016/j.ejps.2024.106999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Thermosensitive liposomes (TSLs) have great potential for the selective delivery of cytostatic drugs to the tumor site with greatly reduced side effects. Here we report the discovery and characterization of new thermosensitive small multilamellar lipid nanoparticles (tSMLPs) with unusually high temperature selectivity. Furthermore, the temperature-dependent release of the fluorescent marker calcein from tSMLPs is enhanced by human serum albumin. tSMLPs can easily be prepared through dual centrifugation (DC) at very high lipid concentrations using dipalmitoyl and distearoyl phosphatidylcholine (DPPC, DSPC) and the phospholipid dipalmitoyl-sn-glycero-phosphatidyldiglycerol (DPPG2). The new particles have a hydrodynamic diameter of about 175 nm and a narrow size distribution (PDI 0.02). tSMLPs consist of multiple lipid membranes, which become increasingly closer packed towards the particle center, and have no visible aqueous core. The particles are highly stable due to strong hydrogen bond-based membrane interactions mediated by DPPG2. tSMLPs can be used as carriers for water-soluble drugs (EE 25 %) entrapped within the interlamellar spaces. Based on biophysical (DSC, DLS and ITC) and morphological (cryo-EM) studies, a hypothesis is presented to explain the structural basis underlying the high temperature selectivity, as well as the unusual morphology of the new thermosensitive lipid nanoparticles.
Collapse
Affiliation(s)
- Jonas K Koehler
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany.
| | - Stefanie Schmager
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Johannes Schnur
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Lars Gedda
- Department of Chemistry-Ångström, Uppsala University, 75237 Uppsala, Sweden
| | - Katarina Edwards
- Department of Chemistry-Ångström, Uppsala University, 75237 Uppsala, Sweden
| | - Heiko Heerklotz
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; Signaling Research Centers BIOSS and CIBBS, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Ulrich Massing
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg im Breisgau, Germany; Andreas Hettich GmbH, 78532 Tuttlingen, Germany.
| |
Collapse
|
9
|
Zhao Z, Fetse J, Mamani UF, Guo Y, Li Y, Patel P, Liu Y, Lin CY, Li Y, Mustafa B, Cheng K. Development of a peptide-based tumor-activated checkpoint inhibitor for cancer immunotherapy. Acta Biomater 2024:S1742-7061(24)00769-4. [PMID: 39716541 DOI: 10.1016/j.actbio.2024.12.051] [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: 08/26/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Abstract
Antibody-based checkpoint inhibitors have achieved great success in cancer immunotherapy, but their uncontrollable immune-related adverse events remain a major challenge. In this study, we developed a tumor-activated nanoparticle that is specifically active in tumors but not in normal tissues. We discovered a short anti-PD-L1 peptide that blocks the PD-1/PD-L1 interaction. The peptide was modified with a PEG chain through a novel matrix metalloproteinase-2 (MMP-2)-specific cleavage linker. The modified TR3 peptide self-assembles into a micelle-like nanoparticle (TR3-M-NP), which remains inactive and unable to block the PD-1/PD-L1 interaction in its native form. However, upon cleavage by MMP-2 in tumors, it releases the active peptide. The TR3-M-NP5k nanoparticle was specifically activated in tumors through enzyme-mediated cleavage, leading to the inhibition of tumor growth and extended survival compared to control groups. In summary, TR3-M-NP shows great potential as a tumor-responsive immunotherapy agent with reduced toxicities. STATEMENT OF SIGNIFICANCE: In this study, we developed a bioactive peptide-based checkpoint inhibitor that is active only in tumors and not in normal tissues, thereby potentially avoiding immune-related adverse effects. We discovered a short anti-PD-L1 peptide, TR3, that blocks the PD-1/PD-L1 interaction. We chemically modified the TR3 peptide to self-assemble into a micelle-like nanoparticle (TR3-M-NP), which itself cannot block the PD-1/PD-L1 interaction but releases the active TR3 peptide in tumors upon cleavage by MMP-2. In contrast, the nanoparticle is randomly degraded in normal tissues into peptides fragments that cannot block the PD-1/PD-L1 interaction. Upon intraperitoneal injection, TR3-M-NP5k was activated specifically in tumors through enzyme cleavage, leading to the inhibition of tumor growth and extended survival compared to the control groups. In summary, TR3-M-NP holds significant promise as a tumor-responsive immunotherapy agent with reduced toxicities. The bioactive platform has the potential to be used for other types of checkpoint inhibitor.
Collapse
Affiliation(s)
- Zhen Zhao
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - John Fetse
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Umar-Farouk Mamani
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yuhan Guo
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yuanke Li
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Pratikkumar Patel
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Chien-Yu Lin
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yongren Li
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Bahaa Mustafa
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA.
| |
Collapse
|
10
|
Wen J, Lei C, Hua S, Cai L, Dai H, Liu S, Li Y, Ivanovski S, Xu C. Regulation of macrophage uptake through the bio-nano interaction using surface functionalized mesoporous silica nanoparticles with large radial pores. J Mater Chem B 2024; 13:137-150. [PMID: 39575665 DOI: 10.1039/d4tb01124d] [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: 12/06/2024]
Abstract
Porous nanoparticles, such as mesoporous silica nanoparticles (MSNs), have garnered significant interest for biomedical applications. Recently, MSNs with large radial pores have attracted increased attention because their unique pore structure and large pore size are suitable for delivering large molecules such as proteins and genes. Upon entry into biological systems like the bloodstream, nanoparticles quickly form a 'protein corona,' leading to alterations in their interactions with immune cells. In this study, we investigated the formation of protein corona on MSNs with large radial pores and various surface modifications using mass spectrometry. We also examined the effects of protein corona on the interaction between MSNs and macrophages. We prepared MSNs with large, cone-shaped radial pores (>30 nm) and six different functional groups, resulting in nanoparticles with neutral, negative, and positive surface charges. Our findings indicate that surface functional groups significantly alter the composition of the protein corona, affecting the bio-nano interaction of these surface-modified MSNs with macrophages. Notably, nanoparticles with similar surface charges exhibited distinct corona characteristics and were internalized differently by macrophages. This underscores the crucial role of the protein corona in determining the fate, behavior, and biological responses of nanoparticles. Our research sheds light on the significance of understanding and controlling protein corona formation to optimize the design and functionality of nanoparticle-based biomedical applications.
Collapse
Affiliation(s)
- Juan Wen
- School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia.
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia
| | - Chang Lei
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Shu Hua
- School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia.
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia
| | - Larry Cai
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Huan Dai
- School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia.
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia
| | - Siyuan Liu
- School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia.
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia
- Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Yiwei Li
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Saso Ivanovski
- School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia.
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia.
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland 4006, Australia
- Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
11
|
Toyoda M, Miura Y, Kobayashi M, Tsuda M, Nomoto T, Honda Y, Nakamura H, Takemoto H, Nishiyama N. Synthesis and Optimization of Ethylenediamine-Based Zwitterion on Polymer Side Chain for Recognizing Narrow Tumorous pH Windows. Biomacromolecules 2024; 25:7788-7798. [PMID: 39481019 PMCID: PMC11632656 DOI: 10.1021/acs.biomac.4c01086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Polyzwitterions that show the alternation of net charge in response to external stimuli have attracted great attention as a new class of surface-polymers on nanomedicines. However, the correlation between their detailed molecular structures and expression of antifouling properties under physiological condition remain controversial. Herein, we synthesized a series of ethylenediamine-based polyzwitterions with carboxy groups/sulfonic groups and ethylene, propylene, and butylene spacers as potential surface-polymers for nanomedicines, allowing sensitive recognition of tumor acidic environments (pH = 6.5-5.5). Then, we evaluated their structure-based characteristics, including pH-dependent cellular uptakes and intracellular distributions. Additionally, the role of conformation stability, i.e., Gibbs free energy changes, was to induce an intramolecular electrostatic interaction in the zwitterionic moieties. These results highlight the practicality of fine-tuning the design of zwitterionic moieties on polymers for the future development of nanomedicines that can recognize the narrow pH window in tumor acidic environments.
Collapse
Affiliation(s)
- Masahiro Toyoda
- Laboratory
for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Department
of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Yutaka Miura
- Laboratory
for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Department
of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Innovation
Center of Nanomedicine (iCONM), Kawasaki
Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Motoaki Kobayashi
- Laboratory
for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Masato Tsuda
- Laboratory
for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Department
of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Takahiro Nomoto
- Department
of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Yuto Honda
- Laboratory
for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Innovation
Center of Nanomedicine (iCONM), Kawasaki
Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Hiroyuki Nakamura
- Laboratory
for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Department
of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Hiroyasu Takemoto
- Medical
Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Nobuhiro Nishiyama
- Laboratory
for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Department
of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Innovation
Center of Nanomedicine (iCONM), Kawasaki
Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| |
Collapse
|
12
|
El-Saadony MT, Fang G, Yan S, Alkafaas SS, El Nasharty MA, Khedr SA, Hussien AM, Ghosh S, Dladla M, Elkafas SS, Ibrahim EH, Salem HM, Mosa WFA, Ahmed AE, Mohammed DM, Korma SA, El-Tarabily MK, Saad AM, El-Tarabily KA, AbuQamar SF. Green Synthesis of Zinc Oxide Nanoparticles: Preparation, Characterization, and Biomedical Applications - A Review. Int J Nanomedicine 2024; 19:12889-12937. [PMID: 39651353 PMCID: PMC11624689 DOI: 10.2147/ijn.s487188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/17/2024] [Indexed: 12/11/2024] Open
Abstract
Over the last decade, biomedical nanomaterials have garnered significant attention due to their remarkable biological properties and diverse applications in biomedicine. Metal oxide nanoparticles (NPs) are particularly notable for their wide range of medicinal uses, including antibacterial, anticancer, biosensing, cell imaging, and drug/gene delivery. Among these, zinc oxide (ZnO) NPs stand out for their versatility and effectiveness. Recently, ZnO NPs have become a primary material in various sectors, such as pharmaceutical, cosmetic, antimicrobials, construction, textile, and automotive industries. ZnO NPs can generate reactive oxygen species and induce cellular apoptosis, thus underpinning their potent anticancer and antibacterial properties. To meet the growing demand, numerous synthetic approaches have been developed to produce ZnO NPs. However, traditional manufacturing processes often involve significant economic and environmental costs, prompting a search for more sustainable alternatives. Intriguingly, biological synthesis methods utilizing plants, plant extracts, or microorganisms have emerged as ideal for producing ZnO NPs. These green production techniques offer numerous medicinal, economic, environmental, and health benefits. This review highlights the latest advancements in the green synthesis of ZnO NPs and their biomedical applications, showcasing their potential to revolutionize the field with eco-friendly and cost-effective solutions.
Collapse
Affiliation(s)
- Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Guihong Fang
- School of Public Health, Heinz Mehlhorn Academician Workstation, Hainan Medical University, Haikou, Hainan, 571199, People’s Republic of China
- Qionghai People’s Hospital, Qionghai, Hainan, 571400, People’s Republic of China
| | - Si Yan
- Qionghai People’s Hospital, Qionghai, Hainan, 571400, People’s Republic of China
| | - Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mahmoud A El Nasharty
- Department of Chemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Sohila A Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta, 31733, Egypt
| | - Aya Misbah Hussien
- Biotechnology Department at Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21531, Egypt
| | - Soumya Ghosh
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Oman
| | - Mthokozisi Dladla
- Human Molecular Biology Unit (School of Biomedical Sciences), Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Shebin El Kom, Menofia, 32511, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Essam H Ibrahim
- Biology Department, Faculty of Science, King Khalid University, Abha, 61413, Saudi Arabia
- Blood Products Quality Control and Research Department, National Organization for Research and Control of Biologicals, Cairo, 12611, Egypt
| | - Heba Mohammed Salem
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Walid F A Mosa
- Plant Production Department (Horticulture-Pomology), Faculty of Agriculture, Saba Basha, Alexandria University, Alexandria, 21531, Egypt
| | - Ahmed Ezzat Ahmed
- Biology Department, Faculty of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Sameh A Korma
- Department of Food Science, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | | | - Ahmed M Saad
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| | - Synan F AbuQamar
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| |
Collapse
|
13
|
Rahat I, Yadav P, Singhal A, Fareed M, Purushothaman JR, Aslam M, Balaji R, Patil-Shinde S, Rizwanullah M. Polymer lipid hybrid nanoparticles for phytochemical delivery: challenges, progress, and future prospects. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:1473-1497. [PMID: 39600519 PMCID: PMC11590012 DOI: 10.3762/bjnano.15.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Phytochemicals, naturally occurring compounds in plants, possess a wide range of therapeutic properties, including antioxidant, anti-inflammatory, anticancer, and antimicrobial activities. However, their clinical application is often hindered by poor water solubility, low bioavailability, rapid metabolism, and instability under physiological conditions. Polymer lipid hybrid nanoparticles (PLHNPs) have emerged as a novel delivery system that combines the advantages of both polymeric and lipid-based nanoparticles to overcome these challenges. This review explores the potential of PLHNPs to enhance the delivery and efficacy of phytochemicals for biomedical applications. We discuss the obstacles in the conventional delivery of phytochemicals, the fundamental architecture of PLHNPs, and the types of PLHNPs, highlighting their ability to improve encapsulation efficiency, stability, and controlled release of the encapsulated phytochemicals. In addition, the surface modification strategies to improve overall therapeutic efficacy by site-specific delivery of encapsulated phytochemicals are also discussed. Furthermore, we extensively discuss the preclinical studies on phytochemical encapsulated PLHNPs for the management of different diseases. Additionally, we explore the challenges ahead and prospects of PLHNPs regarding their widespread use in clinical settings. Overall, PLHNPs hold strong potential for the effective delivery of phytochemicals for biomedical applications. As per the findings from pre-clinical studies, this may offer a promising strategy for managing various diseases.
Collapse
Affiliation(s)
- Iqra Rahat
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut-250005, Uttar Pradesh, India
| | - Pooja Yadav
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut-250005, Uttar Pradesh, India
| | - Aditi Singhal
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut-250005, Uttar Pradesh, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Jaganathan Raja Purushothaman
- Department of Orthopaedics, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai-602105, Tamil Nadu, India
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001, Kurdistan Region, Iraq
| | - Raju Balaji
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai-602105, Tamil Nadu, India
| | - Sonali Patil-Shinde
- Department of Pharmaceutical Chemistry, Dr. D.Y Patil Institute of Pharmaceutical Sciences and Research, Pimpri Pune-411018, Maharashtra, India
| | - Md Rizwanullah
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| |
Collapse
|
14
|
Su Q, Chen J, Liu Z, Fan Y, He S. A pH-Sensitive cRGD-PEG-siRNA Conjugated Compound Targeting Glioblastoma. Bioconjug Chem 2024; 35:1732-1743. [PMID: 39431993 PMCID: PMC11583972 DOI: 10.1021/acs.bioconjchem.4c00255] [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: 10/22/2024]
Abstract
Glioblastoma ranks among the most prevalent primary intracranial tumors, characterized by high mortality and poor prognosis. Chemotherapy remains a key treatment strategy for gliomas, though most current drugs suffer from limited efficacy and significant toxicity. This study focuses on a cRGD-siEGFR coupling compound synthesized in a previous stage. Prior research indicated that cRGD-siEGFR molecules exhibited certain targeting and antitumor properties but faced issues of inadequate targeting, low efficacy, and high renal toxicity. To enhance antitumor efficacy and mitigate side effects, a pH-responsive, long-circulating, and highly targeted siRNA delivery system, the cRGD-PEG-siEGFR conjugate, was developed. The targeting, antitumor effects, and biological distribution of cRGD-PEG-siEGFR were examined. The results demonstrated that cRGD-PEG-siEGFR was effectively taken up by αvβ3-positive U87MG cells, specifically silenced EGFR gene expression, and exhibited antitumor effects. In normal physiological conditions, it avoided uptake by normal cells, thereby reducing side effects. Furthermore, in vivo biodistribution experiments revealed that cRGD-PEG-siEGFR, compared to cRGD-siEGFR, significantly decreased renal accumulation and exhibited prolonged circulation. Consequently, cRGD-PEG-siRNA emerges as a promising drug candidate with attributes of long circulation, high targeting, pH responsiveness, and substantial antitumor efficacy.
Collapse
Affiliation(s)
- Qing Su
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510655, China
| | - Junxiao Chen
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510655, China
| | - Ziyuan Liu
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510655, China
| | - Yiqi Fan
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510655, China
| | - Shuai He
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510655, China
| |
Collapse
|
15
|
Han S, Yoo W, Carton O, Joo J, Kwon EJ. PEGylated Multimeric RNA Nanoparticles for siRNA Delivery in Traumatic Brain Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2405806. [PMID: 39498752 DOI: 10.1002/smll.202405806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/27/2024] [Indexed: 11/07/2024]
Abstract
Traumatic brain injury (TBI) impacts millions of people globally, however currently there are no approved therapeutics that address long-term brain health. In order to create a technology that is relevant for siRNA delivery in TBI after systemic administration, sub-100 nm nanoparticles with rolling circle transcription (RCT) are synthesized and isolated in order improve payload delivery into the injured brain. Unlike conventional RCT-based RNA particles, in this method, sub-100 nm RNA nanoparticles (RNPs) are isolated. To enhance RNP pharmacokinetics, RNPs are synthesized with modified bases in order to graft polyethylene glycol (PEG) to the RNPs. PEGylated RNPs (PEG-RNPs) do not significantly impact their knockdown activity in vitro and lead to longer blood half-life after systemic administration and greater accumulation into the injured brain in a mouse model of TBI. In order to demonstrate RNA interference (RNAi) activity of RNPs, knockdown of the inflammatory cytokine TNF-α in injured brain tissue after systemic administration of RNPs in a mouse model of TBI is demonstrated. In summary, small sub-100 nm multimeric RNA nanoparticles are synthesized and isolated that can be modified using accessible chemistry in order to create a technology suitable for systemic RNAi therapy for TBI.
Collapse
Affiliation(s)
- Sangwoo Han
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Woojung Yoo
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Olivia Carton
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- Graduate School of Health Science and Technology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Republic of Korea
- Materials Research Science and Engineering Center, 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
- Materials Research Science and Engineering Center, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| |
Collapse
|
16
|
He P, Zou M, Zhang C, Shi Y, Qin L. Celastrol-Loaded Hyaluronic Acid/Cancer Cell Membrane Lipid Nanoparticles for Targeted Hepatocellular Carcinoma Prevention. Cells 2024; 13:1819. [PMID: 39513925 PMCID: PMC11545145 DOI: 10.3390/cells13211819] [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: 09/21/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide, and its prevention and treatment face severe challenges. It is crucial to improve the targeting of drugs on tumor cells and tissues. Celastrol (CeT), as an active ingredient of traditional Chinese medicine, possesses strong antitumor effects, especially in triggering apoptosis of HCC. However, due to its toxicity and lack of targeting, its application is greatly limited. HMCLPs, a nano-biomimetic platform carrying CeT with controllable drug release, enhanced targeting, and immunocompatibility, were developed for the first time, which can be used for the treatment of HCC. By utilizing homologous cell membranes and hyaluronic acid (HA), HMCLPs can precisely target tumor regions and release CeT in a controlled manner. Both in vitro and in vivo studies have demonstrated that HMCLPs loaded with CeT significantly increased the accumulation of reactive oxygen species (ROS), induced mitochondrial damage, and triggered apoptosis of HCC cells, resulting in effective treatment with minimal adverse reaction. The development of HMCLPs as a nanocarrier system for CeT delivery offers a promising therapeutic strategy for HCC. This innovative approach improves the targeted delivery and bioavailability of CeT, dramatically induces apoptosis in HCC cells, and exerts its powerful antitumor effects while minimizing systemic toxicity. The present study highlights the potential of combining innovative nanocarriers with powerful natural compounds such as CeT to enhance efficacy and reduce toxicity.
Collapse
Affiliation(s)
- Peng He
- Laboratory of Stem Cell Regulation and Application of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (P.H.); (C.Z.); (Y.S.)
| | - Manshu Zou
- Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha 410208, China;
| | - Chanjuan Zhang
- Laboratory of Stem Cell Regulation and Application of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (P.H.); (C.Z.); (Y.S.)
| | - Yaning Shi
- Laboratory of Stem Cell Regulation and Application of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (P.H.); (C.Z.); (Y.S.)
| | - Li Qin
- Laboratory of Stem Cell Regulation and Application of Traditional Chinese Medicine, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (P.H.); (C.Z.); (Y.S.)
| |
Collapse
|
17
|
Barros AS, Pinto S, Viegas J, Martins C, Almeida H, Alves I, Pinho S, Nunes R, Harris A, Sarmento B. Orally Delivered Stimulus-Sensitive Nanomedicine to Harness Teduglutide Efficacy in Inflammatory Bowel Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402502. [PMID: 39007246 DOI: 10.1002/smll.202402502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/03/2024] [Indexed: 07/16/2024]
Abstract
Inflammatory Bowel Disease (IBD) is a chronic inflammatory condition affecting the gastrointestinal tract (GIT). Glucagon-like peptide-2 (GLP-2) analogs possess high potential in the treatment of IBD by enhancing intestinal repair and attenuating inflammation. Due to the enzymatic degradation and poor intestinal absorption, GLP-2 analogs are administered parenterally, which leads to poor patient compliance. This work aims to develop IBD-targeted nanoparticles (NPs) for the oral delivery of the GLP-2 analog, Teduglutide (TED). Leveraging the overproduction of Reactive Oxygen Species (ROS) in the IBD environment, ROS-sensitive NPs are developed to target the intestinal epithelium, bypassing the mucus barrier. PEGylation of NPs facilitates mucus transposition, but subsequent PEG removal is crucial for cellular internalization. This de-PEGylation is possible by including a ROS-sensitive thioketal linker within the system. ROS-sensitive NPs are established, with the ability to fully de-PEGylate via ROS-mediated cleavage. Encapsulation of TED into NPs resulted in the absence of absorption in 3D in vitro models, potentially promoting a localized action, and avoiding adverse effects due to systemic absorption. Upon oral administration to colitis-induced mice, ROS-sensitive NPs are located in the colon, displaying healing capacity and reducing inflammation. Cleavable PEGylated NPs demonstrate effective potential in managing IBD symptoms and modulating the disease's progression.
Collapse
Affiliation(s)
- Andreia S Barros
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4050-313, Portugal
| | - Soraia Pinto
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4050-313, Portugal
| | - Juliana Viegas
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
| | - Claúdia Martins
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
| | - Helena Almeida
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4050-313, Portugal
| | - Inês Alves
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
| | - Salomé Pinho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- ICBAS- Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, 4050-313, Portugal
- FMUP- Faculty of Medicine, University of Porto, Porto, 4200-319, Portugal
| | - Rute Nunes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- IUCS-CESPU-Instituto Universitário de Ciências das Saúde, Porto, 4585-116, Portugal
| | - Alan Harris
- Ferring Pharmaceuticals, 1162-Saint-Prex, SA Chemin de la Vergognausaz 50, Switzerland
| | - Bruno Sarmento
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- IUCS-CESPU-Instituto Universitário de Ciências das Saúde, Porto, 4585-116, Portugal
| |
Collapse
|
18
|
Zhao H, Zhang C, Tian C, Li L, Wu B, Stuart MAC, Wang M, Zhou X, Wang J. Rational design of diblock copolymer enables efficient cytosolic protein delivery. J Colloid Interface Sci 2024; 673:722-734. [PMID: 38901362 DOI: 10.1016/j.jcis.2024.06.123] [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: 04/11/2024] [Revised: 06/03/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
Polymer-mediated cytosolic protein delivery demonstrates a promising strategy for the development of protein therapeutics. Here, we propose a new designed diblock copolymer which realizes efficient cytosolic protein delivery both in vitro and in vivo. The polymer contains one protein-binding block composed of phenylboronic acid (PBA) and N-(3-dimethylaminopropyl) (DMAP) pendant units for protein binding and endosomal escape, respectively, followed by the response to ATP enriched in the cytosol which triggers the protein release. The other block is PEG designed to improve particle size control and circulation in vivo. By optimizing the block composition, sequence and length of the copolymer, the optimal one (BP20) was identified with the binding block containing 20 units of both PBA and DMAP, randomly distributed along the chain. When mixed with proteins, the BP20 forms stable nanoparticles and mediates efficient cytosolic delivery of a wide range of proteins including enzymes, toxic proteins and CRISPR/Cas9 ribonucleoproteins (RNP), to various cell lines. The PEG block, especially when further modified with folic acid (FA), enables tumor-targeted delivery of Saporin in vivo, which significantly suppresses the tumor growth. Our results shall inspire the design of novel polymeric vehicles with robust capability for cytosolic protein delivery, which holds great potential for both biological research and therapeutic applications.
Collapse
Affiliation(s)
- Hongyang Zhao
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, People's Republic of China
| | - Chenglin Zhang
- Department of Orthopedics, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, 200003 Shanghai, People's Republic of China
| | - Chang Tian
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, People's Republic of China
| | - Lingshu Li
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, People's Republic of China
| | - Bohang Wu
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, People's Republic of China
| | - Martien A Cohen Stuart
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, People's Republic of China
| | - Mingwei Wang
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, People's Republic of China.
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, 200003 Shanghai, People's Republic of China.
| | - Junyou Wang
- State-Key Laboratory of Chemical Engineering, and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, 130 Meilong Road, 200237 Shanghai, People's Republic of China.
| |
Collapse
|
19
|
Zhao L, Feng L, Shan R, Huang Y, Shen L, Fan M, Wang Y. Nanoparticle-based approaches for treating restenosis after vascular injury. Front Pharmacol 2024; 15:1427651. [PMID: 39512830 PMCID: PMC11540800 DOI: 10.3389/fphar.2024.1427651] [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: 05/04/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
Percutaneous coronary intervention (PCI) is currently the main method for treating coronary artery stenosis, but the incidence of restenosis after PCI is relatively high. Restenosis, the narrowing of blood vessels by more than 50% of the normal diameter after PCI, severely compromises the therapeutic efficacy. Therefore, preventing postinterventional restenosis is important. Vascular restenosis is mainly associated with endothelial injury, the inflammatory response, the proliferation and migration of vascular smooth muscle cells (VSMCs), excessive deposition of extracellular matrix (ECM) and intimal hyperplasia (IH) and is usually prevented by administering antiproliferative or anti-inflammatory drugs through drug-eluting stents (DESs); however, DESs can lead to uncontrolled drug release. In addition, as extracorporeal implants, they can cause inflammation and thrombosis, resulting in suboptimal treatment. Therefore, there is an urgent need for a drug carrier with controlled drug release and high biocompatibility for in vivo drug delivery to prevent restenosis. The development of nanotechnology has enabled the preparation of nanoparticle drug carriers with low toxicity, high drug loading, high biocompatibility, precise targeting, controlled drug release and excellent intracellular delivery ability. This review summarizes the advantages of nanoparticle drug carriers for treating vascular restenosis, as well as how nanoparticles have improved targeting, slowed the release of therapeutic agents, and prolonged circulation in vivo to prevent vascular restenosis more effectively. The overall purpose of this review is to present an overview of nanoparticle therapy for vascular restenosis. We expect these findings to provide insight into nanoparticle-based therapeutic approaches for vascular restenosis.
Collapse
Affiliation(s)
- Liangfeng Zhao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Liuliu Feng
- Department of Cardiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Rong Shan
- Department of Cardiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Yue Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Mingliang Fan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yu Wang
- Department of Cardiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
20
|
Haseeb M, Khan I, Kartal Z, Mahfooz S, Hatiboglu MA. Status Quo in the Liposome-Based Therapeutic Strategies Against Glioblastoma: "Targeting the Tumor and Tumor Microenvironment". Int J Mol Sci 2024; 25:11271. [PMID: 39457052 PMCID: PMC11509082 DOI: 10.3390/ijms252011271] [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: 09/14/2024] [Revised: 10/06/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Glioblastoma is the most aggressive and fatal brain cancer, characterized by a high growth rate, invasiveness, and treatment resistance. The presence of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) poses a challenging task for chemotherapeutics, resulting in low efficacy, bioavailability, and increased dose-associated side effects. Despite the rigorous treatment strategies, including surgical resection, radiotherapy, and adjuvant chemotherapy with temozolomide, overall survival remains poor. The failure of current chemotherapeutics and other treatment regimens in glioblastoma necessitates the development of new drug delivery methodologies to precisely and efficiently target glioblastoma. Nanoparticle-based drug delivery systems offer a better therapeutic option in glioblastoma, considering their small size, ease of diffusion, and ability to cross the BBB. Liposomes are a specific category of nanoparticles made up of fatty acids. Furthermore, liposomes can be surface-modified to target a particular receptor and are nontoxic. This review discusses various methods of liposome modification for active/directed targeting and various liposome-based therapeutic approaches in the delivery of current chemotherapeutic drugs and nucleic acids in targeting the glioblastoma and tumor microenvironment.
Collapse
Affiliation(s)
- Mohd Haseeb
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
| | - Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zeynep Kartal
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, 34820 Istanbul, Turkey; (M.H.); (S.M.)
- Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, 34093 Istanbul, Turkey
| |
Collapse
|
21
|
Digiacomo L, Renzi S, Pirrottina A, Amenitsch H, De Lorenzi V, Pozzi D, Cardarelli F, Caracciolo G. PEGylation-Dependent Cell Uptake of Lipid Nanoparticles Revealed by Spatiotemporal Correlation Spectroscopy. ACS Pharmacol Transl Sci 2024; 7:3004-3010. [PMID: 39421655 PMCID: PMC11480925 DOI: 10.1021/acsptsci.4c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Polyethylene glycol (PEG) is a common surface modification for lipid nanoparticles (LNPs) to improve their stability and in vivo circulation time. However, the impact of PEGylation on LNP cellular uptake remains poorly understood. To tackle this issue, we systematically compared plain and PEGylated LNPs by combining dynamic light scattering, electrophoretic light scattering, and synchrotron small-angle X-ray scattering (SAXS) that unveils a striking similarity in size and core structure but a significant reduction in surface charge. Upon administration to human embryonic kidney (HEK 293) cells, plain and PEGylated LNPs were internalized through different endocytic routes, as revealed by spatiotemporal correlation spectroscopy. An imaging-derived mean square displacement (iMSD) analysis shows that PEGylated LNPs exhibit a significantly stronger preference for caveolae-mediated endocytosis (CAV) and clathrin-mediated endocytosis (CME) pathways compared to plain LNPs, with these latter being better tailored to MCR-dependent internalization and trafficking. This suggests that PEG plays a crucial role in directing LNPs toward specific cellular uptake routes. Further studies should explore how PEG-mediated endocytosis impacts intracellular trafficking and ultimately translates to therapeutic efficacy, guiding the design of next-generation LNP delivery systems.
Collapse
Affiliation(s)
- Luca Digiacomo
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, 00161 Rome, Italy
| | - Serena Renzi
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, 00161 Rome, Italy
| | - Andrea Pirrottina
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, 00161 Rome, Italy
| | - Heinz Amenitsch
- Institute
of Inorganic Chemistry, Graz University
of Technology, 8010 Graz, Austria
| | | | - Daniela Pozzi
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, 00161 Rome, Italy
| | | | - Giulio Caracciolo
- NanoDelivery
Lab, Department of Molecular Medicine, Sapienza
University of Rome, 00161 Rome, Italy
| |
Collapse
|
22
|
Qi Y, Han H, Liu A, Zhao S, Lawanprasert A, Nielsen JE, Choudhary H, Liang D, Barron AE, Murthy N. Ethylene oxide graft copolymers reduce the immunogenicity of lipid nanoparticles. RSC Adv 2024; 14:30071-30076. [PMID: 39309654 PMCID: PMC11414743 DOI: 10.1039/d4ra05007j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Lipid nanoparticle (LNP)/mRNA complexes have great therapeutic potential but their PEG chains can induce the production of anti-PEG antibodies. New LNPs that do not contain PEG are greatly needed. We demonstrate here that poly-glutamic acid-ethylene oxide graft copolymers can replace the PEG on LNPs and outperform PEG-LNPs after chronic administration.
Collapse
Affiliation(s)
- Yalin Qi
- Department of Bioengineering, University of California, Berkeley Berkeley California 94720 USA
- Innovative Genomics Institute (IGI) Berkeley California 94704 USA
| | - Hesong Han
- Department of Bioengineering, University of California, Berkeley Berkeley California 94720 USA
- Innovative Genomics Institute (IGI) Berkeley California 94704 USA
| | - Albert Liu
- Department of Bioengineering, University of California, Berkeley Berkeley California 94720 USA
- Innovative Genomics Institute (IGI) Berkeley California 94704 USA
| | - Sheng Zhao
- Department of Bioengineering, University of California, Berkeley Berkeley California 94720 USA
- Innovative Genomics Institute (IGI) Berkeley California 94704 USA
| | - Atip Lawanprasert
- Department of Bioengineering, University of California, Berkeley Berkeley California 94720 USA
- Innovative Genomics Institute (IGI) Berkeley California 94704 USA
| | - Josefine Eilsø Nielsen
- Department of Bioengineering, School of Medicine, Stanford University Stanford California 94305 USA
- Department of Science and Environment, Roskilde University Roskilde 4000 Denmark
| | - Hema Choudhary
- Department of Bioengineering, University of California, Berkeley Berkeley California 94720 USA
- Innovative Genomics Institute (IGI) Berkeley California 94704 USA
| | - Dengpan Liang
- Department of Bioengineering, University of California, Berkeley Berkeley California 94720 USA
- Innovative Genomics Institute (IGI) Berkeley California 94704 USA
| | - Annelise E Barron
- Department of Bioengineering, School of Medicine, Stanford University Stanford California 94305 USA
| | - Niren Murthy
- Department of Bioengineering, University of California, Berkeley Berkeley California 94720 USA
- Innovative Genomics Institute (IGI) Berkeley California 94704 USA
| |
Collapse
|
23
|
Wan Q, Yuan H, Cai P, Liu Y, Yan T, Wang L, Zhou Z, Zhang W, Liu N. Effects of PEGylation on Imaging Contrast of 68Ga-Labeled Bicyclic Peptide PET Probes Targeting Nectin-4. Mol Pharm 2024; 21:4430-4440. [PMID: 39069891 DOI: 10.1021/acs.molpharmaceut.4c00366] [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] [Indexed: 07/30/2024]
Abstract
Nectin cell adhesion molecule 4 (Nectin-4) is overexpressed in various malignant tumors and has emerged as a promising target for tumor imaging. Bicyclic peptides, known for their conformational rigidity, metabolic stability, and membrane permeability, are ideal tracers for positron emission tomography (PET) imaging. In this study, we evaluated the feasibility of visualizing Nectin-4-positive tumors using radiolabeled bicyclic peptide derivatives and optimized the pharmacokinetics of radiotracers by introducing PEG chains of different lengths. Five PEGylated radiotracers radiolabeled with 68Ga3+ exhibited high radiochemical purity and stability. As the chain length increased, the Log D values decreased from -2.32 ± 0.13 to -2.50 ± 0.16, indicating a gradual increase in the hydrophilicity of the radiotracers. In vitro cell-binding assay results showed that the PEGylated bicyclic peptide exhibits nanomolar affinity, and blocking experiments confirmed the specific binding of the tracers to the Nectin-4 receptor. In vivo PET imaging and biodistribution studies in SW780 and 5637 xenograft mice showed that [68Ga]Ga-NOTA-PEG12-BP demonstrated optimal pharmacokinetics, characterized by rapid and good tumor uptake, faster background clearance, and improved tumor-to-tissue contrast. Finally, compared with 18F-FDG, PET imaging, in vivo blocking assays of [68Ga]Ga-NOTA-PEG12-BP and histological staining confirmed that specific tumor uptake was mediated by Nectin-4 receptors. The results indicated that [68Ga]Ga-NOTA-PEG12-BP was a promising PET radiotracer for Nectin-4 targeting, with applications for clinical translation.
Collapse
Affiliation(s)
- Qiang Wan
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
| | - Hongmei Yuan
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
| | - Ping Cai
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Yang Liu
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
| | - Ting Yan
- Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Li Wang
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
| | - Zhijun Zhou
- Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Department of Nuclear Medicine, The Affiliated Hospital, Southwest Medical University, Jiangyang District, Luzhou 646000, Sichuan, China
| | - Wei Zhang
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Nan Liu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| |
Collapse
|
24
|
Grzetic DJ, Hamilton NB, Shelley JC. Coarse-Grained Simulation of mRNA-Loaded Lipid Nanoparticle Self-Assembly. Mol Pharm 2024; 21:4747-4753. [PMID: 39145436 DOI: 10.1021/acs.molpharmaceut.4c00216] [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] [Indexed: 08/16/2024]
Abstract
Ionizable lipid-containing lipid nanoparticles (LNPs) have enabled the delivery of RNA for a range of therapeutic applications. In order to optimize safe, targeted, and effective LNP-based RNA delivery platforms, an understanding of the role of composition and pH in their structural properties and self-assembly is crucial, yet there have been few computational studies of such phenomena. Here we present a coarse-grained model of ionizable lipid and mRNA-containing LNPs. Our model allows access to the large length- and time-scales necessary for LNP self-assembly and is mapped and parametrized with reference to all-atom structures and simulations of the corresponding components at compositions typical of LNPs used for mRNA delivery. Our simulations reveal insights into the dynamics of self-assembly of such mRNA-encapsulating LNPs, as well as the subsequent pH change-driven LNP morphology and release of mRNA.
Collapse
Affiliation(s)
- Douglas J Grzetic
- Schrödinger, Inc., 101 SW Main Street, Suite 1300, Portland, Oregon 97204, United States
| | - Nicholas B Hamilton
- Schrödinger, Inc., 101 SW Main Street, Suite 1300, Portland, Oregon 97204, United States
- Department of Chemistry, University of Vermont, Burlington, Vermont 05405, United States
| | - John C Shelley
- Schrödinger, Inc., 101 SW Main Street, Suite 1300, Portland, Oregon 97204, United States
| |
Collapse
|
25
|
Shchegravina ES, Tretiakova DS, Sitdikova AR, Usova SD, Boldyrev IA, Alekseeva AS, Svirshchevskaya EV, Vodovozova EL, Fedorov AY. Design and preparation of pH-sensitive cytotoxic liposomal formulations containing antitumor colchicine analogues for target release. J Liposome Res 2024; 34:399-410. [PMID: 37867342 DOI: 10.1080/08982104.2023.2274428] [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: 08/19/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Herein, we describe the synthesis of pH-sensitive lipophilic colchicine prodrugs for liposomal bilayer inclusion, as well as preparation and characterization of presumably stealth PEGylated liposomes with above-mentioned prodrugs. These formulations liberate strongly cytotoxic colchicinoid derivatives selectively under slightly acidic tumor-associated conditions, ensuring tumor-targeted delivery of the compounds. The design of the prodrugs is addressed to pH-triggered release of active compounds in the slight acidic media, that corresponds to tumor microenvironment, while keeping sufficient stability of the whole formulation at physiological pH. Correlations between the structure of the conjugates, their hydrolytic stability, colloidal stability, ability of the prodrug retention in the lipid bilayer are described. Several formulations were found promising for further development and in vivo investigations.
Collapse
Affiliation(s)
- Ekaterina S Shchegravina
- Department of Organic Chemistry, UNN Lobachevsky University, Nizhny Novgorod, Russian Federation
| | - Daria S Tretiakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russian Federation
| | - Alsu R Sitdikova
- Department of Organic Chemistry, UNN Lobachevsky University, Nizhny Novgorod, Russian Federation
| | - Sofia D Usova
- N.D. Zelinsky Insitute of Organic Chemistry RAS, Moscow, Russian Federation
| | - Ivan A Boldyrev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russian Federation
| | - Anna S Alekseeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russian Federation
| | | | - Elena L Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russian Federation
| | - Alexey Yu Fedorov
- Department of Organic Chemistry, UNN Lobachevsky University, Nizhny Novgorod, Russian Federation
| |
Collapse
|
26
|
García-Fernández C, Virgilio T, Latino I, Guerra-Rebollo M, F Gonzalez S, Borrós S, Fornaguera C. Stealth mRNA nanovaccines to control lymph node trafficking. J Control Release 2024; 374:325-336. [PMID: 39154934 DOI: 10.1016/j.jconrel.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/15/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
mRNA-based vaccines symbolize a new paradigm shift in personalized medicine for the treatment of infectious and non-infectious diseases. However, the reactogenicity associated with the currently approved formulations limits their applicability in autoinflammatory disorders, such as tumour therapeutics. In this study, we present a delivery system showing controlled immunogenicity and minimal non-specific inflammation, allowing for selective delivery of mRNA to antigen presenting cells (APCs) within the medullary region of the lymph nodes. Our platform offers precise control over the trafficking of nanoparticles within the lymph nodes by optimizing stealth and targeting properties, as well as the subsequent opsonization process. By targeting specific cells, we observed a potent adaptive and humoral immune response, which holds promise for preventive and therapeutic anti-tumoral vaccines. Through spatial programming of nanoparticle distribution, we can promote robust immunization, thus improving and expanding the utilization of mRNA vaccines. This innovative approach signifies a remarkable step forward in the field of targeted nanomedicine.
Collapse
Affiliation(s)
- Coral García-Fernández
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL). Via Augusta, Barcelona, Catalonia, 08017, Spain; Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Universita della Svitzzera italiana (USI) - Switzerland, Via Francesco Chiesa 5, Bellinzona 6500, Suiza
| | - Tommaso Virgilio
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Universita della Svitzzera italiana (USI) - Switzerland, Via Francesco Chiesa 5, Bellinzona 6500, Suiza
| | - Irene Latino
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Universita della Svitzzera italiana (USI) - Switzerland, Via Francesco Chiesa 5, Bellinzona 6500, Suiza
| | - Marta Guerra-Rebollo
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL). Via Augusta, Barcelona, Catalonia, 08017, Spain
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Universita della Svitzzera italiana (USI) - Switzerland, Via Francesco Chiesa 5, Bellinzona 6500, Suiza
| | - Salvador Borrós
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL). Via Augusta, Barcelona, Catalonia, 08017, Spain
| | - Cristina Fornaguera
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL). Via Augusta, Barcelona, Catalonia, 08017, Spain.
| |
Collapse
|
27
|
Zhang L, Li Y, Liu X, He X, Zhang J, Zhou J, Qiao Y, Wu H, Sun F, Zhou Q. Optimal development of apoptotic cells-mimicking liposomes targeting macrophages. J Nanobiotechnology 2024; 22:501. [PMID: 39169328 PMCID: PMC11337832 DOI: 10.1186/s12951-024-02755-3] [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: 03/12/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Macrophages are multifunctional innate immune cells that play indispensable roles in homeostasis, tissue repair, and immune regulation. However, dysregulated activation of macrophages is implicated in the pathogenesis of various human disorders, making them a potential target for treatment. Through the expression of pattern recognition and scavenger receptors, macrophages exhibit selective uptake of pathogens and apoptotic cells. Consequently, the utilization of drug carriers that mimic pathogenic or apoptotic signals shows potential for targeted delivery to macrophages. In this study, a series of mannosylated or/and phosphatidylserine (PS) -presenting liposomes were developed to target macrophages via the design of experiment (DoE) strategy and the trial-and-error (TaE) approach. The optimal molar ratio for the liposome formulation was DOPC: DSPS: Chol: PEG-PE = 20:60:20:2 based on the results of cellular uptake and cytotoxicity evaluation on RAW 264.7 and THP-1 in vitro. Results from in vivo distribution showed that, in the DSS-induced colitis model and collagen II-induced rheumatoid arthritis model, PS-presenting liposomes (PS-Lipo) showed the highest accumulation in intestine and paws respectively, which holds promising potential for macrophage target therapy since macrophages are abundant at inflammatory sites and contribute to the progression of corresponding diseases. Organs such as the heart, liver, spleen, lung, and kidney did not exhibit histological alterations such as inflammation or necrosis when exposed to PC-presenting liposomes (PC-Lipo) or PS-Lipo. In addition, liposomes demonstrated hemobiocompatibility and no toxicity to liver or kidney for circulation and did not induce metabolic injury in the animals. Thus, the well-designed PS-Lipo demonstrated the most potential for macrophage target therapy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Prosthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School,Institute of Stomatology,Nanjing University, Nanjing, 210002, China
| | - Yujiao Li
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xing Liu
- Department of Infectious Disease and Liver Disease, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Xiaolu He
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Jieyu Zhang
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Jun Zhou
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Youbei Qiao
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China
| | - Hong Wu
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, Xi'an, 710032, China.
| | - Fangfang Sun
- Department of Prosthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School,Institute of Stomatology,Nanjing University, Nanjing, 210002, China.
| | - Qing Zhou
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
28
|
Du Q, Liu Y, Fan M, Wei S, Ismail M, Zheng M. PEG length effect of peptide-functional liposome for blood brain barrier (BBB) penetration and brain targeting. J Control Release 2024; 372:85-94. [PMID: 38838784 DOI: 10.1016/j.jconrel.2024.06.005] [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: 03/04/2024] [Revised: 05/21/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Nanoparticles, in particular PEGylated, show great potential for in vivo brain targeted drug delivery. Nevertheless, how polyethylene glycol (PEG) length of nanoparticles affects their blood brain barrier (BBB) penetration or brain targeting is still unclear. In this study, we investigated the power of PEG chain-lengths (2, 3.4, 5, 10 kDa) in BBB penetration and brain targeting using Angiopep-2 peptide decorated liposomes. We found that PEG chain-length is critical, where the shorter PEG enabled the Angiopep-2 decorated liposomes to display more potent in vitro cell uptake via endocytosis. In contrast, their in vitro BBB penetration via transcytosis was much weaker relative to the liposomes with longer PEG chains, which result from their ineffective BBB exocytosis. Interestingly, the in vivo brain targeting aligns with the in vitro BBB penetration, as the long chain PEG-modified liposomes exerted superior brain accumulation both in normal or orthotropic glioblastoma (GBM) bearing mice, which could be ascribed to the combinational effect of prolonged circulation and enhanced BBB penetration of long chain PEG attached liposomes. These results demonstrate the crucial role of PEG length of nanoparticles for BBB penetration and brain targeting, providing guidance for PEG length selection in the design of nanocarrier for brain diseases treatment.
Collapse
Affiliation(s)
- Qiuli Du
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yang Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Mengyu Fan
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Sijun Wei
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Muhammad Ismail
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, Henan International Joint Laboratory of Nanobiomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
29
|
Lisiecka MZ. Polyethylene glycol and immunology: aspects of allergic reactions and their mechanisms, as well as ways to prevent them in clinical practice. Immunol Res 2024; 72:675-682. [PMID: 38502278 DOI: 10.1007/s12026-024-09473-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/14/2024] [Indexed: 03/21/2024]
Abstract
In modern medical practice, where polyethylene glycol is widely used as a component of various drugs, such as vaccines, chemotherapy drugs, and antibiotics, including vaccines, the issue of allergic reactions to this substance is becoming increasingly important. The purpose of this study is to review and systematise data on various aspects of allergic reactions to polyethylene glycol with the aim of better understanding their pathogenesis, clinical manifestations, diagnostic methods, and possible treatment approaches. The study analysed literature data in modern databases, such as MEDLINE, PubMed, and Scopus, on allergic reactions to polyethylene glycol, using the keywords: "PEG", "polyethylene glycol", "allergy", "side effect". The main aspects of allergy to this substance were highlighted, including mechanisms of development, diagnostic methods, and possible treatment strategies. The analysis found that allergic reactions to polyethylene glycol can manifest in a variety of ways, including anaphylaxis and systemic reactions. A possible role for the immune response has been identified, including the production of IgE and IgM antibodies, complement activation, and accelerated clearance in response to polyethylene glycol, in blood plasma. Data are also provided on how to diagnose an increased risk of an allergic reaction in patients who have previously received drugs with this type of drug transporter and in patients receiving high molecular weight types of polyethylene glycol. The results of this review contribute to a better understanding of allergic reactions to polyethylene glycol and provide information for the development of more effective diagnostic and treatment methods.
Collapse
Affiliation(s)
- Maria Zofia Lisiecka
- Department of Allergology, National Medical Institute of the Ministry of the Interior and Administration, 137 Woloska Str, 02-507, Warsaw, Poland.
| |
Collapse
|
30
|
Sardo C, Auriemma G, Mazzacano C, Conte C, Piccolo V, Ciaglia T, Denel-Bobrowska M, Olejniczak AB, Fiore D, Proto MC, Gazzerro P, Aquino RP. Inulin Amphiphilic Copolymer-Based Drug Delivery: Unraveling the Structural Features of Graft Constructs. Pharmaceutics 2024; 16:971. [PMID: 39204316 PMCID: PMC11359108 DOI: 10.3390/pharmaceutics16080971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
In this study, the structural attributes of nanoparticles obtained by a renewable and non-immunogenic "inulinated" analog of the "pegylated" PLA (PEG-PLA) were examined, together with the potential of these novel nanocarriers in delivering poorly water-soluble drugs. Characterization of INU-PLA assemblies, encompassing critical aggregation concentration (CAC), NMR, DLS, LDE, and SEM analyses, was conducted to elucidate the core/shell architecture of the carriers and in vitro cyto- and hemo-compatibility were assayed. The entrapment and in vitro delivery of sorafenib tosylate (ST) were also studied. INU-PLA copolymers exhibit distinctive features: (1) Crew-cut aggregates are formed with coronas of 2-4 nm; (2) a threshold surface density of 1 INU/nm2 triggers a configuration change; (3) INU surface density influences PLA core dynamics, with hydrophilic segment stretching affecting PLA distribution towards the interface. INU-PLA2NPs demonstrated an outstanding loading of ST and excellent biological profile, with effective internalization and ST delivery to HepG2 cells, yielding a comparable IC50.
Collapse
Affiliation(s)
- Carla Sardo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.A.); (C.M.); (T.C.); (D.F.); (M.C.P.); (P.G.); (R.P.A.)
| | - Giulia Auriemma
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.A.); (C.M.); (T.C.); (D.F.); (M.C.P.); (P.G.); (R.P.A.)
| | - Carmela Mazzacano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.A.); (C.M.); (T.C.); (D.F.); (M.C.P.); (P.G.); (R.P.A.)
| | - Claudia Conte
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (C.C.)
| | - Virgilio Piccolo
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy; (C.C.)
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.A.); (C.M.); (T.C.); (D.F.); (M.C.P.); (P.G.); (R.P.A.)
| | - Marta Denel-Bobrowska
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland; (M.D.-B.); (A.B.O.)
| | - Agnieszka B. Olejniczak
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland; (M.D.-B.); (A.B.O.)
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.A.); (C.M.); (T.C.); (D.F.); (M.C.P.); (P.G.); (R.P.A.)
| | - Maria Chiara Proto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.A.); (C.M.); (T.C.); (D.F.); (M.C.P.); (P.G.); (R.P.A.)
| | - Patrizia Gazzerro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.A.); (C.M.); (T.C.); (D.F.); (M.C.P.); (P.G.); (R.P.A.)
| | - Rita Patrizia Aquino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (G.A.); (C.M.); (T.C.); (D.F.); (M.C.P.); (P.G.); (R.P.A.)
| |
Collapse
|
31
|
Cheng Z, Fobian SF, Gurrieri E, Amin M, D'Agostino VG, Falahati M, Zalba S, Debets R, Garrido MJ, Saeed M, Seynhaeve ALB, Balcioglu HE, Ten Hagen TLM. Lipid-based nanosystems: the next generation of cancer immune therapy. J Hematol Oncol 2024; 17:53. [PMID: 39030582 PMCID: PMC11265205 DOI: 10.1186/s13045-024-01574-1] [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: 05/23/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Immunotherapy has become an important part of the oncotherapy arsenal. Its applicability in various cancer types is impressive, as well as its use of endogenous mechanisms to achieve desired ends. However, off-target or on-target-off-tumor toxicity, limited activity, lack of control in combination treatments and, especially for solid tumors, low local accumulation, have collectively limited clinical use thereof. These limitations are partially alleviated by delivery systems. Lipid-based nanoparticles (NPs) have emerged as revolutionary carriers due to favorable physicochemical characteristics, with specific applications and strengths particularly useful in immunotherapeutic agent delivery. The aim of this review is to highlight the challenges faced by immunotherapy and how lipid-based NPs have been, and may be further utilized to address such challenges. We discuss recent fundamental and clinical applications of NPs in a range of areas and provide a detailed discussion of the main obstacles in immune checkpoint inhibition therapies, adoptive cellular therapies, and cytokine therapies. We highlight how lipid-based nanosystems could address these through either delivery, direct modulation of the immune system, or targeting of the immunosuppressive tumor microenvironment. We explore advanced and emerging liposomal and lipid nanoparticle (LNP) systems for nucleic acid delivery, intrinsic and extrinsic stimulus-responsive formulations, and biomimetic lipid-based nanosystems in immunotherapy. Finally, we discuss the key challenges relating to the clinical use of lipid-based NP immunotherapies, suggesting future research directions for the near term to realize the potential of these innovative lipid-based nanosystems, as they become the crucial steppingstone towards the necessary enhancement of the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Ziyun Cheng
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Seth-Frerich Fobian
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Elena Gurrieri
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Mohamadreza Amin
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vito Giuseppe D'Agostino
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Mojtaba Falahati
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sara Zalba
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - María J Garrido
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarra Institute for Health Research, Pamplona, Spain
| | - Mesha Saeed
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ann L B Seynhaeve
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hayri E Balcioglu
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands.
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
32
|
Hao T, Zhang B, Li W, Yang X, Wu S, Yuan Y, Cui H, Chen Q, Li Z. Nordihydroguaiaretic Acid-Cross-Linked Phenylboronic Acid-Functionalized Polyplex Micelles for Anti-angiogenic Gene Therapy of Orthotopic and Metastatic Tumors. ACS APPLIED MATERIALS & INTERFACES 2024; 16:34620-34631. [PMID: 38934519 DOI: 10.1021/acsami.4c05311] [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: 06/28/2024]
Abstract
Polyplexes are required to be equipped with multiple functionalities to accomplish adequate structure stability and gene transfection efficacy for gene therapy. Herein, a 4-carboxy-3-fluorophenylboronic acid (FPBA)-functionalized block copolymer of PEG-b-PAsp(DET/FBA) and PAsp(DET/FBA) (abbreviated as PB and HB) was synthesized and applied for engineering functional polyplex micelles (PMs) through ionic complexation with pDNA followed by strategic cross-linking with nordihydroguaiaretic acid (NDGA) in respect to the potential linkage of polyphenol and FPBA moieties. In relation to polyplex micelles void of cross-linking, the engineered multifunctional polyplex micelles (PBHBN-PMs) were determined to possess improved structural tolerability against the exchange reaction with charged species. Besides, the FPBA/NDGA cross-linking appeared to be selectively cleaved in the acidic endosomal compartments but not the neutral milieu. Furthermore, the PBHB-PMs with the optimal FPBA/NDGA cross-linking degree were identified to possess appreciable cellular uptake and endosomal escape activities, eliciting a significantly high level of gene expression relative to P-PMs and PB-PMs. Eventually, in vivo antitumor therapy by our proposed multifunctional PMs appeared to be capable of facilitating expression of the antiangiogenic genomic payloads (sFlt-1 pDNA) via systemic administration. The enriched antiangiogenic sFlt-1 in the tumors could silence the activities of angiogenic cytokines for the inhibited neo-vasculature and the suppressed growth of orthotopic 4T1 tumors. Of note, the persistent expression of the antiangiogenic sFlt-1 is also presumed to migrate into the blood circulation, thereby accounting for an overall antiangiogenic environment in preventing the potential pulmonary metastasis. Hence, our elaborated multifaceted PMs inspired fascinating potential as an intriguing gene delivery system for the treatment of clinical solid tumors and metastasis.
Collapse
Affiliation(s)
- Tangna Hao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Bingning Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Wenjing Li
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Xianxian Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Sha Wu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yujie Yuan
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Hongxia Cui
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Qixian Chen
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Zhen Li
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| |
Collapse
|
33
|
Li H, Du Z, Zhu L, Zhang C, Xiong J, Zhou B, Dong B, Zhang X, Alifu N. Ultrabright NIR-IIb Fluorescence Quantum Dots for Targeted Imaging-Guided Surgery. ACS APPLIED MATERIALS & INTERFACES 2024; 16:32045-32057. [PMID: 38861701 DOI: 10.1021/acsami.4c04748] [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: 06/13/2024]
Abstract
Pioneering approaches for precise tumor removal involve fluorescence-guided surgery, while challenges persist, including the low fluorescence contrast observed at tumor boundaries and the potential for excessive damage to normal tissue at the edges. Lead/cadmium sulfide quantum dots (PbS@CdS QDs), boasting high quantum yields (QYs) and vivid fluorescence, have facilitated advancements in the second near-infrared window (NIR-II, 900-1700 nm). However, during fluorescent surgical navigation operations, hydrophilic coatings of these inorganic nanoparticles (NPs) guarantee biosafety; it also comes at the expense of losing a significant portion of QY and NIR-II fluorescence, causing heightened damage to normal tissues caused by cutting edges. Herein, we present hydrophilic core-shell PbS@CdS@PEG NPs with an exceptionally small diameter (∼8 nm) and a brilliant NIR-IIb (1500-1700 nm) emission at approximately 1600 nm. The mPEG-SH (MW: 2000) addresses the hydrophobicity and enhances the biosafety of PbS@CdS QDs. In vivo fluorescence-guided cervical tumor resection becomes achievable immediately upon injection of an aqueous solution of PbS@CdS@PEG NPs. Notably, this approach results in a significantly reduced thickness (100-500 μm) of damage to normal tissues at the margins of the resected tumors. With a high QY (∼30.2%) and robust resistance to photobleaching, NIR-IIb imaging is sustained throughout the imaging process.
Collapse
Affiliation(s)
- Hui Li
- Department of Epidemiology and Health Statistics, School of Public Health, Xinjiang Medical University, Urumqi 830054, China
| | - Zhong Du
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 841100, China
| | - Lijun Zhu
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 841100, China
| | - Chi Zhang
- Department of Labor Hygiene and Environmental Hygiene, School of Public Health, Xinjiang Medical University, Urumqi 830054, China
| | - Jiabao Xiong
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 841100, China
| | - Bingshuai Zhou
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Biao Dong
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology, Xinjiang Medical University, Urumqi 830054, China
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Xueliang Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Xinjiang Medical University, Urumqi 830054, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology, Xinjiang Medical University, Urumqi 830054, China
| | - Nuernisha Alifu
- Department of Epidemiology and Health Statistics, School of Public Health, Xinjiang Medical University, Urumqi 830054, China
- Department of Labor Hygiene and Environmental Hygiene, School of Public Health, Xinjiang Medical University, Urumqi 830054, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology, Xinjiang Medical University, Urumqi 830054, China
- The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 841100, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| |
Collapse
|
34
|
Susa F, Arpicco S, Pirri CF, Limongi T. An Overview on the Physiopathology of the Blood-Brain Barrier and the Lipid-Based Nanocarriers for Central Nervous System Delivery. Pharmaceutics 2024; 16:849. [PMID: 39065547 PMCID: PMC11279990 DOI: 10.3390/pharmaceutics16070849] [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: 05/11/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The state of well-being and health of our body is regulated by the fine osmotic and biochemical balance established between the cells of the different tissues, organs, and systems. Specific districts of the human body are defined, kept in the correct state of functioning, and, therefore, protected from exogenous or endogenous insults of both mechanical, physical, and biological nature by the presence of different barrier systems. In addition to the placental barrier, which even acts as a linker between two different organisms, the mother and the fetus, all human body barriers, including the blood-brain barrier (BBB), blood-retinal barrier, blood-nerve barrier, blood-lymph barrier, and blood-cerebrospinal fluid barrier, operate to maintain the physiological homeostasis within tissues and organs. From a pharmaceutical point of view, the most challenging is undoubtedly the BBB, since its presence notably complicates the treatment of brain disorders. BBB action can impair the delivery of chemical drugs and biopharmaceuticals into the brain, reducing their therapeutic efficacy and/or increasing their unwanted bioaccumulation in the surrounding healthy tissues. Recent nanotechnological innovation provides advanced biomaterials and ad hoc customized engineering and functionalization methods able to assist in brain-targeted drug delivery. In this context, lipid nanocarriers, including both synthetic (liposomes, solid lipid nanoparticles, nanoemulsions, nanostructured lipid carriers, niosomes, proniosomes, and cubosomes) and cell-derived ones (extracellular vesicles and cell membrane-derived nanocarriers), are considered one of the most successful brain delivery systems due to their reasonable biocompatibility and ability to cross the BBB. This review aims to provide a complete and up-to-date point of view on the efficacy of the most varied lipid carriers, whether FDA-approved, involved in clinical trials, or used in in vitro or in vivo studies, for the treatment of inflammatory, cancerous, or infectious brain diseases.
Collapse
Affiliation(s)
- Francesca Susa
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| | - Candido Fabrizio Pirri
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Tania Limongi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| |
Collapse
|
35
|
Saadh MJ, Mustafa MA, Kumar A, Alamir HTA, Kumar A, Khudair SA, Faisal A, Alubiady MHS, Jalal SS, Shafik SS, Ahmad I, Khry FAF, Abosaoda MK. Stealth Nanocarriers in Cancer Therapy: a Comprehensive Review of Design, Functionality, and Clinical Applications. AAPS PharmSciTech 2024; 25:140. [PMID: 38890191 DOI: 10.1208/s12249-024-02843-5] [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: 02/17/2024] [Accepted: 05/13/2024] [Indexed: 06/20/2024] Open
Abstract
Nanotechnology has significantly transformed cancer treatment by introducing innovative methods for delivering drugs effectively. This literature review provided an in-depth analysis of the role of nanocarriers in cancer therapy, with a particular focus on the critical concept of the 'stealth effect.' The stealth effect refers to the ability of nanocarriers to evade the immune system and overcome physiological barriers. The review investigated the design and composition of various nanocarriers, such as liposomes, micelles, and inorganic nanoparticles, highlighting the importance of surface modifications and functionalization. The complex interaction between the immune system, opsonization, phagocytosis, and the protein corona was examined to understand the stealth effect. The review carefully evaluated strategies to enhance the stealth effect, including surface coating with polymers, biomimetic camouflage, and targeting ligands. The in vivo behavior of stealth nanocarriers and their impact on pharmacokinetics, biodistribution, and toxicity were also systematically examined. Additionally, the review presented clinical applications, case studies of approved nanocarrier-based cancer therapies, and emerging formulations in clinical trials. Future directions and obstacles in the field, such as advancements in nanocarrier engineering, personalized nanomedicine, regulatory considerations, and ethical implications, were discussed in detail. The review concluded by summarizing key findings and emphasizing the transformative potential of stealth nanocarriers in revolutionizing cancer therapy. This review enhanced the comprehension of nanocarrier-based cancer therapies and their potential impact by providing insights into advanced studies, clinical applications, and regulatory considerations.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, University of Imam Jaafar AL-Sadiq, Baghdad, Iraq
| | - Ashwani Kumar
- Department of Life Sciences, School of Sciences, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Pharmacy, Vivekananda Global University, Jaipur, Rajasthan, India
| | | | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, 247341, Uttar Pradesh, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | | | - Ahmed Faisal
- Department of Pharmacy, Al-Noor University College, Nineveh, Iraq
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Shafik Shaker Shafik
- Experimental Nuclear Radiation Group, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Faeza A F Khry
- Faculty of pharmacy, department of pharmaceutics, Al-Esraa University, Baghdad, Iraq
| | - Munther Kadhim Abosaoda
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Qadisiyyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
36
|
Guido V, Olivieri PH, Brito ML, Prezoto BC, Martinez DST, Oliva MLV, Sousa AA. Stealth and Biocompatible Gold Nanoparticles through Surface Coating with a Zwitterionic Derivative of Glutathione. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:12167-12178. [PMID: 38808371 PMCID: PMC11171461 DOI: 10.1021/acs.langmuir.4c01123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Gold nanoparticles (AuNPs) hold promise in biomedicine, but challenges like aggregation, protein corona formation, and insufficient biocompatibility must be thoroughly addressed before advancing their clinical applications. Designing AuNPs with specific protein corona compositions is challenging, and strategies for corona elimination, such as coating with polyethylene glycol (PEG), have limitations. In this study, we introduce a commercially available zwitterionic derivative of glutathione, glutathione monoethyl ester (GSHzwt), for the surface coating of colloidal AuNPs. Particles coated with GSHzwt were investigated alongside four other AuNPs coated with various ligands, including citrate ions, tiopronin, glutathione, cysteine, and PEG. We then undertook a head-to-head comparison of these AuNPs to assess their behavior in biological fluid. GSHzwt-coated AuNPs exhibited exceptional resistance to aggregation and protein adsorption. The particles could also be readily functionalized with biotin and interact with streptavidin receptors in human plasma. Additionally, they exhibited significant blood compatibility and noncytotoxicity. In conclusion, GSHzwt provides a practical and easy method for the surface passivation of AuNPs, creating "stealth" particles for potential clinical applications.
Collapse
Affiliation(s)
- Vinicius
S. Guido
- Department
of Biochemistry, Federal University of São
Paulo, São
Paulo 04044-020, Brazil
| | - Paulo H. Olivieri
- Department
of Biochemistry, Federal University of São
Paulo, São
Paulo 04044-020, Brazil
| | - Milena L. Brito
- Brazilian
Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo 13083-100, Brazil
| | - Benedito C. Prezoto
- Laboratory
of Pharmacology, the Butantan Institute, São Paulo 05503-900, Brazil
| | - Diego S. T. Martinez
- Brazilian
Nanotechnology National Laboratory (LNNano), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo 13083-100, Brazil
| | - Maria Luiza V. Oliva
- Department
of Biochemistry, Federal University of São
Paulo, São
Paulo 04044-020, Brazil
| | - Alioscka A. Sousa
- Department
of Biochemistry, Federal University of São
Paulo, São
Paulo 04044-020, Brazil
| |
Collapse
|
37
|
Şahin Z, Önal E, Ali LMA, Durand D, Emami A, Touré M, İşci U, Gary-Bobo M, Cammas-Marion S, Dumoulin F. Nanoencapsulation of a Far-Red Absorbing Phthalocyanine into Poly(benzylmalate) Biopolymers and Modulation of Their Photodynamic Efficiency. Biomacromolecules 2024; 25:3261-3270. [PMID: 38752976 PMCID: PMC11170942 DOI: 10.1021/acs.biomac.3c01382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 06/11/2024]
Abstract
Two different poly(benzylmalate) biopolymers, a hydrophobic non-PEGylated (PMLABe73) and an amphiphilic PEGylated derivative (PEG42-b-PMLABe73), have been used to encapsulate a phthalocyanine chosen for its substitution pattern that is highly suitable for photodynamic therapy. Different phthalocyanine/(co)polymers ratios have been used for the nanoprecipitation. A set of six nanoparticles has been obtained. If the amphiphilic PEGylated copolymer proved to be slightly more efficient for the encapsulation and to lower the aggregation of the phthalocyanine inside the nanoparticles, it is, however, the hydrophobic PMLABe73-based nanoparticles that exhibited the best photodynamic efficiency.
Collapse
Affiliation(s)
- Zeynel Şahin
- Faculty
of Technology, Department of Metallurgical & Materials Engineering, Marmara University, 34722 Istanbul, Türkiye
| | - Emel Önal
- Faculty
of Engineering, Doğuş University, Ümraniye, 34775 Istanbul, Türkiye
| | - Lamiaa M. A. Ali
- IBMM,
Univ Montpellier, CNRS, ENSCM, 34093 Montpellier, France
- Department
of Biochemistry Medical Research Institute, University of Alexandria, 21561 Alexandria, Egypt
| | - Denis Durand
- IBMM,
Univ Montpellier, CNRS, ENSCM, 34093 Montpellier, France
| | - Atefeh Emami
- Faculty
of Engineering and Natural Sciences, Biomedical Engineering Department, Acıbadem Mehmet Ali Aydınlar University, Ataşehir, 34752 Istanbul, Türkiye
| | - Marie Touré
- IBMM,
Univ Montpellier, CNRS, ENSCM, 34093 Montpellier, France
| | - Umit İşci
- Faculty
of Technology, Department of Metallurgical & Materials Engineering, Marmara University, 34722 Istanbul, Türkiye
| | | | - Sandrine Cammas-Marion
- Univ
Rennes,
ENSCR, INSA Rennes, CNRS, ISCR (Institut des Sciences Chimiques de
Rennes)—UMR 6226, F-35000 Rennes, France
- INSERM,
INRAE, Univ Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer),
U1317, F-35000 Rennes, France
| | - Fabienne Dumoulin
- Faculty
of Engineering and Natural Sciences, Biomedical Engineering Department, Acıbadem Mehmet Ali Aydınlar University, Ataşehir, 34752 Istanbul, Türkiye
| |
Collapse
|
38
|
Miatmoko A, Octavia RT, Araki T, Annoura T, Sari R. Advancing liposome technology for innovative strategies against malaria. Saudi Pharm J 2024; 32:102085. [PMID: 38690211 PMCID: PMC11059525 DOI: 10.1016/j.jsps.2024.102085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
This review discusses the potential of liposomes as drug delivery systems for antimalarial therapies. Malaria continues to be a significant cause of mortality and morbidity, particularly among children and pregnant women. Drug resistance due to patient non-compliance and troublesome side effects remains a significant challenge in antimalarial treatment. Liposomes, as targeted and efficient drug carriers, have garnered attention owing to their ability to address these issues. Liposomes encapsulate hydrophilic and/or hydrophobic drugs, thus providing comprehensive and suitable therapeutic drug delivery. Moreover, the potential of passive and active drug delivery enables drug concentration in specific target tissues while reducing adverse effects. However, successful liposome formulation is influenced by various factors, including drug physicochemical characteristics and physiological barriers encountered during drug delivery. To overcome these challenges, researchers have explored modifications in liposome nanocarriers to achieve efficient drug loading, controlled release, and system stability. Computational approaches have also been adopted to predict liposome system stability, membrane integrity, and drug-liposome interactions, improving formulation development efficiency. By leveraging computational methods, optimizing liposomal drug delivery systems holds promise for enhancing treatment efficacy and minimizing side effects in malaria therapy. This review consolidates the current understanding and highlights the potential of liposome strategies against malaria.
Collapse
Affiliation(s)
- Andang Miatmoko
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
- Stem Cell Research and Development Center, Universitas Airlangga, 2 Floor Institute of Tropical Disease Building, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
- Nanotechnology and Drug Delivery System Research Group, Faculty of Pharmacy, Universitas Airlangga, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
| | - Rifda Tarimi Octavia
- Master Program of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
| | - Tamasa Araki
- Department of Parasitology, National Institute of Infectious Diseases (NIID), 1-23-1 Toyama, Shinju-ku, Tokyo 162-8640, Japan
| | - Takeshi Annoura
- Department of Parasitology, National Institute of Infectious Diseases (NIID), 1-23-1 Toyama, Shinju-ku, Tokyo 162-8640, Japan
| | - Retno Sari
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Campus C UNAIR Mulyorejo, Surabaya 60115, Indonesia
| |
Collapse
|
39
|
Liau B, Zhang L, Ang MJY, Ng JY, C V SB, Schneider S, Gudihal R, Bae KH, Yang YY. Quantitative analysis of mRNA-lipid nanoparticle stability in human plasma and serum by size-exclusion chromatography coupled with dual-angle light scattering. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 58:102745. [PMID: 38499167 DOI: 10.1016/j.nano.2024.102745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/20/2024]
Abstract
Understanding the stability of mRNA loaded lipid nanoparticles (mRNA-LNPs) is imperative for their clinical development. Herein, we propose the use of size-exclusion chromatography coupled with dual-angle light scattering (SEC-MALS) as a new approach to assessing mRNA-LNP stability in pure human serum and plasma. By applying a dual-column configuration to attenuate interference from plasma components, SEC-MALS was able to elucidate the degradation kinetics and physical property changes of mRNA-LNPs, which have not been observed accurately by conventional dynamic light scattering techniques. Interestingly, both serum and plasma had significantly different impacts on the molecular weight and radius of gyration of mRNA-LNPs, suggesting the involvement of clotting factors in desorption of lipids from mRNA-LNPs. We also discovered that a trace impurity (~1 %) in ALC-0315, identified as its O-tert-butyloxycarbonyl-protected form, greatly diminished mRNA-LNP stability in serum. These results demonstrated the potential utility of SEC-MALS for optimization and quality control of LNP formulations.
Collapse
Affiliation(s)
- Brian Liau
- Agilent Technologies, 1 Yishun Avenue 7, Singapore 768923, Republic of Singapore.
| | - Li Zhang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Republic of Singapore
| | - Melgious Jin Yan Ang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Republic of Singapore
| | - Jian Yao Ng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Republic of Singapore
| | - Suresh Babu C V
- Agilent Technologies, 1 Yishun Avenue 7, Singapore 768923, Republic of Singapore
| | - Sonja Schneider
- Agilent Technologies Deutschland GmbH, Hewlett-Packard Strasse 8, 76337 Waldbronn, Germany
| | - Ravindra Gudihal
- Agilent Technologies, 1 Yishun Avenue 7, Singapore 768923, Republic of Singapore
| | - Ki Hyun Bae
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Republic of Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Republic of Singapore.
| |
Collapse
|
40
|
Andrade-Gagnon B, Casillas-Popova SN, Jazani AM, Oh JK. Design, Synthesis, and Acid-Responsive Disassembly of Shell-Sheddable Block Copolymer Labeled with Benzaldehyde Acetal Junction. Macromol Rapid Commun 2024; 45:e2400097. [PMID: 38499007 DOI: 10.1002/marc.202400097] [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: 02/15/2024] [Revised: 03/13/2024] [Indexed: 03/20/2024]
Abstract
Smart nanoassemblies degradable through the cleavage of acid-labile linkages have attracted significant attention because of their biological relevance found in tumor tissues. Despite their high potential to achieve controlled/enhanced drug release, a systematic understanding of structural factors that affect their pH sensitivity remains challenging, particulary in the consruction of effective acid-degradable shell-sheddable nanoassemblies. Herein, the authors report the synthesis and acid-responsive degradation through acid-catalyzed hydrolysis of three acetal and ketal diols and identify benzaldehyde acetal (BzAA) exhibiting optimal hydrolysis profiles in targeted pH ranges to be a suitable candidate for junction acid-labile linkage. The authors explore the synthesis and aqueous micellization of well-defined poly(ethylene glycol)-based block copolymer bearing BzAA linkage covalently attached to a polymethacrylate block for the formation of colloidally-stable nanoassemblies with BzAA groups at core/corona interfaces. Promisingly, the investigation on acid-catalyzed hydrolysis and disassembly shows that the formed nanoassemblies meet the criteria for acid-degradable shell-sheddable nanoassemblies: slow degradation at tumoral pH = 6.5 and rapid disassembly at endo/lysosomal pH = 5.0, while colloidal stability at physiological pH = 7.4. This work guides the design principle of acid-degradable shell-sheddable nanoassemblies bearing BzAA at interfaces, thus offering the promise to address the PEG dilemma and improve endocytosis in tumor-targeting drug delivery.
Collapse
Affiliation(s)
- Brandon Andrade-Gagnon
- Department of Chemistry and Biochemistry, Concordia University, Montreal, QC, H4B 1R6, Canada
| | | | - Arman Moini Jazani
- Department of Chemistry and Biochemistry, Concordia University, Montreal, QC, H4B 1R6, Canada
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Jung Kwon Oh
- Department of Chemistry and Biochemistry, Concordia University, Montreal, QC, H4B 1R6, Canada
| |
Collapse
|
41
|
Zelepukin IV, Shevchenko KG, Deyev SM. Rediscovery of mononuclear phagocyte system blockade for nanoparticle drug delivery. Nat Commun 2024; 15:4366. [PMID: 38777821 PMCID: PMC11111695 DOI: 10.1038/s41467-024-48838-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
Rapid uptake of nanoparticles by mononuclear phagocyte system (MPS) significantly hampers their therapeutic efficacy. Temporal MPS blockade is one of the few ways to overcome this barrier - the approach rediscovered many times under different names but never extensively used in clinic. Using meta-analysis of the published data we prove the efficacy of this technique for enhancing particle circulation in blood and their delivery to tumours, describe a century of its evolution and potential combined mechanism behind it. Finally, we discuss future directions of the research focusing on the features essential for successful clinical translation of the method.
Collapse
Affiliation(s)
- Ivan V Zelepukin
- Department of Medicinal Chemistry, Uppsala University, 751 23, Uppsala, Sweden.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia.
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997, Moscow, Russia
| |
Collapse
|
42
|
Urbano-Gámez JD, Guzzi C, Bernal M, Solivera J, Martínez-Zubiaurre I, Caro C, García-Martín ML. Tumor versus Tumor Cell Targeting in Metal-Based Nanoparticles for Cancer Theranostics. Int J Mol Sci 2024; 25:5213. [PMID: 38791253 PMCID: PMC11121233 DOI: 10.3390/ijms25105213] [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: 04/11/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The application of metal-based nanoparticles (mNPs) in cancer therapy and diagnostics (theranostics) has been a hot research topic since the early days of nanotechnology, becoming even more relevant in recent years. However, the clinical translation of this technology has been notably poor, with one of the main reasons being a lack of understanding of the disease and conceptual errors in the design of mNPs. Strikingly, throughout the reported studies to date on in vivo experiments, the concepts of "tumor targeting" and "tumor cell targeting" are often intertwined, particularly in the context of active targeting. These misconceptions may lead to design flaws, resulting in failed theranostic strategies. In the context of mNPs, tumor targeting can be described as the process by which mNPs reach the tumor mass (as a tissue), while tumor cell targeting refers to the specific interaction of mNPs with tumor cells once they have reached the tumor tissue. In this review, we conduct a critical analysis of key challenges that must be addressed for the successful targeting of either tumor tissue or cancer cells within the tumor tissue. Additionally, we explore essential features necessary for the smart design of theranostic mNPs, where 'smart design' refers to the process involving advanced consideration of the physicochemical features of the mNPs, targeting motifs, and physiological barriers that must be overcome for successful tumor targeting and/or tumor cell targeting.
Collapse
Affiliation(s)
- Jesús David Urbano-Gámez
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - Cinzia Guzzi
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - Manuel Bernal
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, 29071 Malaga, Spain
| | - Juan Solivera
- Department of Neurosurgery, Reina Sofia University Hospital, 14004 Cordoba, Spain;
| | - Iñigo Martínez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, P.O. Box 6050, Langnes, 9037 Tromsö, Norway;
| | - Carlos Caro
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - María Luisa García-Martín
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
- Biomedical Research Networking Center in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
43
|
Pan F, Liu M, Li G, Chen B, Chu Y, Yang Y, Wu E, Yu Y, Lin S, Ding T, Wei X, Zhan C, Qian J. Phospholipid Type Regulates Protein Corona Composition and In Vivo Performance of Lipid Nanodiscs. Mol Pharm 2024; 21:2272-2283. [PMID: 38607681 DOI: 10.1021/acs.molpharmaceut.3c01084] [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] [Indexed: 04/14/2024]
Abstract
Over the years, there has been significant interest in PEGylated lipid-based nanocarriers within the drug delivery field. The inevitable interplay between the nanocarriers and plasma protein plays a pivotal role in their in vivo biological fate. Understanding the factors influencing lipid-based nanocarrier and protein corona interactions is of paramount importance in the design and clinical translation of these nanocarriers. Herein, discoid-shaped lipid nanodiscs (sNDs) composed of different phospholipids with varied lipid tails and head groups were fabricated. We investigated the impact of phospholipid components on the interaction between sNDs and serum proteins, particle stability, and biodistribution. The results showed that all of these lipid nanodiscs remained stable over a 15 day storage period, while their stability in the blood serum demonstrated significant differences. The sND composed of POPG exhibited the least stability due to its potent complement activation capability, resulting in rapid blood clearance. Furthermore, a negative correlation between the complement activation capability and serum stability was identified. Pharmacokinetic and biodistribution experiments indicated that phospholipid composition did not influence the capability of sNDs to evade the accelerated blood clearance phenomenon. Complement deposition on the sND was inversely associated with the area under the curve. Additionally, all lipid nanodiscs exhibited dominant adsorption of apolipoprotein. Remarkably, the POPC-based lipid nanodisc displayed a significantly higher deposition of apolipoprotein E, contributing to an obvious brain distribution, which provides a promising tool for brain-targeted drug delivery.
Collapse
Affiliation(s)
- Feng Pan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmacy, Jing'an District Central Hospital of Shanghai Fudan University, Shanghai 201203, P. R. China
| | - Mengyuan Liu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmacy, Jing'an District Central Hospital of Shanghai Fudan University, Shanghai 201203, P. R. China
| | - Guanghui Li
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmacy, Jing'an District Central Hospital of Shanghai Fudan University, Shanghai 201203, P. R. China
| | - Boqian Chen
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Yuxiu Chu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Yang Yang
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Ercan Wu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Yifei Yu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Shiqi Lin
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Tianhao Ding
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Xiaoli Wei
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center & Department of Pharmacology School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P. R. China
| | - Jun Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education & Department of Pharmacy, Jing'an District Central Hospital of Shanghai Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
44
|
Zimmer O, Goepferich A. On the uncertainty of the correlation between nanoparticle avidity and biodistribution. Eur J Pharm Biopharm 2024; 198:114240. [PMID: 38437906 DOI: 10.1016/j.ejpb.2024.114240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/05/2024] [Accepted: 02/28/2024] [Indexed: 03/06/2024]
Abstract
The specific delivery of a drug to its site of action also known as targeted drug delivery is a topic in the field of pharmaceutics studied for decades. One approach extensively investigated in this context is the use ligand functionalized nanoparticles. These particles are modified to carry receptor specific ligands, enabling them to accumulate at a desired target site. However, while this concept initially appears straightforward to implement, in-depth research has revealed several challenges hindering target site specific particle accumulation - some of which remain unresolved to this day. One of these challenges consists in the still incomplete understanding of how nanoparticles interact with biological systems. This knowledge gap significantly compromises the predictability of particle distribution in biological systems, which is critical for therapeutic efficacy. One of the most crucial steps in delivery is the attachment of nanoparticles to cells at the target site. This attachment occurs via the formation of multiple ligand receptor bonds. A process also referred to as multivalent interaction. While multivalency has been described extensively for individual molecules and macromolecules respectively, little is known on the multivalent binding of nanoparticles to cells. Here, we will specifically introduce the concept of avidity as a measure for favorable particle membrane interactions. Also, an overview about nanoparticle and membrane properties affecting avidity will be given. Thereafter, we provide a thorough review on literature investigating the correlation between nanoparticle avidity and success in targeted particle delivery. In particular, we want to analyze the currently uncertain data on the existence and nature of the correlation between particle avidity and biodistribution.
Collapse
Affiliation(s)
- Oliver Zimmer
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Bavaria 93053, Germany.
| |
Collapse
|
45
|
Li ZZ, Zhong NN, Cao LM, Cai ZM, Xiao Y, Wang GR, Liu B, Xu C, Bu LL. Nanoparticles Targeting Lymph Nodes for Cancer Immunotherapy: Strategies and Influencing Factors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308731. [PMID: 38327169 DOI: 10.1002/smll.202308731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/07/2024] [Indexed: 02/09/2024]
Abstract
Immunotherapy has emerged as a potent strategy in cancer treatment, with many approved drugs and modalities in the development stages. Despite its promise, immunotherapy is not without its limitations, including side effects and suboptimal efficacy. Using nanoparticles (NPs) as delivery vehicles to target immunotherapy to lymph nodes (LNs) can improve the efficacy of immunotherapy drugs and reduce side effects in patients. In this context, this paper reviews the development of LN-targeted immunotherapeutic NP strategies, the mechanisms of NP transport during LN targeting, and their related biosafety risks. NP targeting of LNs involves either passive targeting, influenced by NP physical properties, or active targeting, facilitated by affinity ligands on NP surfaces, while alternative methods, such as intranodal injection and high endothelial venule (HEV) targeting, have uncertain clinical applicability and require further research and validation. LN targeting of NPs for immunotherapy can reduce side effects and increase biocompatibility, but risks such as toxicity, organ accumulation, and oxidative stress remain, although strategies such as biodegradable biomacromolecules, polyethylene glycol (PEG) coating, and impurity addition can mitigate these risks. Additionally, this work concludes with a future-oriented discussion, offering critical insights into the field.
Collapse
Affiliation(s)
- Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Ze-Min Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Chun Xu
- School of Dentistry, The University of Queensland, 288 Herston Road, Brisbane, 4066, Australia
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| |
Collapse
|
46
|
Wu W, Wu W, Guo M, Wang R, Wang X, Gao Q. Synthesis of MPEG-b-PLLA Diblock Copolymers and Their Crystallization Performance with PDLA and PLLA Composite Films. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2105. [PMID: 38730912 PMCID: PMC11084635 DOI: 10.3390/ma17092105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024]
Abstract
Methoxy poly(ethylene glycol)-block-poly(L-lactide) (MPEG-b-PLLA) has a wide range of applications in pharmaceuticals and biology, and its structure and morphology have been thoroughly studied. In the experiment, we synthesized MPEG-b-PLLA with different block lengths using the principle of ring-opening polymerization by controlling the amount of lactic acid added. The thermodynamic properties of copolymers and the crystallization properties of blends were studied separately. The crystallization kinetics of PDLA/MPEG-b-PLA and PLLA/MPEG-b-PLA composite films were studied using differential scanning calorimetry (DSC). The results indicate that the crystallization kinetics of composite films are closely related to the amount of block addition. The crystallinity of the sample first increases and then decreases with an increase in MPEG-b-PLLA content. These results were also confirmed in polarized optical microscope (POM) and wide-angle X-ray diffraction (WAXD) tests. When 3% MPEG-b-PLLA was added to the PDLA matrix, the blend exhibited the strongest crystallization performance.
Collapse
Affiliation(s)
- Wenjing Wu
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China; (W.W.); (W.W.); (R.W.); (X.W.)
| | - Weixin Wu
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China; (W.W.); (W.W.); (R.W.); (X.W.)
| | - Mingwei Guo
- College of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China;
| | - Ruizhe Wang
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China; (W.W.); (W.W.); (R.W.); (X.W.)
| | - Xuanxuan Wang
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China; (W.W.); (W.W.); (R.W.); (X.W.)
| | - Qinwei Gao
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China; (W.W.); (W.W.); (R.W.); (X.W.)
- Jiangsu Key Laboratory for the Chemistry and Utilization of Agricultural and Forest Biomass, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
47
|
Bento C, Katz M, Santos MMM, Afonso CAM. Striving for Uniformity: A Review on Advances and Challenges To Achieve Uniform Polyethylene Glycol. Org Process Res Dev 2024; 28:860-890. [PMID: 38660381 PMCID: PMC11036406 DOI: 10.1021/acs.oprd.3c00428] [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: 11/05/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 04/26/2024]
Abstract
Poly(ethylene glycol) (PEG) is the polymer of choice in drug delivery systems due to its biocompatibility and hydrophilicity. For over 20 years, this polymer has been widely used in the drug delivery of small drugs, proteins, oligonucleotides, and liposomes, improving the stability and pharmacokinetics of many drugs. However, despite the extensive clinical experience with PEG, concerns have emerged related to its use. These include hypersensitivity, purity, and nonbiodegradability. Moreover, conventional PEG is a mixture of polymers that can complicate drug synthesis and purification leading to unwanted immunogenic reactions. Studies have shown that uniform PEGylated drugs may be more effective than conventional PEGylated drugs as they can overcome issues related to molecular heterogeneity and immunogenicity. This has led to significant research efforts to develop synthetic procedures to produce uniform PEGs (monodisperse PEGs). As a result, iterative step-by-step controlled synthesis methods have been created over time and have shown promising results. Nonetheless, these procedures have presented numerous challenges due to their iterative nature and the requirement for multiple purification steps, resulting in increased costs and time consumption. Despite these challenges, the synthetic procedures went through several improvements. This review summarizes and discusses recent advances in the synthesis of uniform PEGs and its derivatives with a focus on overall yields, scalability, and purity of the polymers. Additionally, the available characterization methods for assessing polymer monodispersity are discussed as well as uniform PEG applications, side effects, and possible alternative polymers that can overcome the drawbacks.
Collapse
Affiliation(s)
- Cláudia Bento
- Hovione
Farmaciência S.A., Estrada do Paço do Lumiar, Campus do Lumiar, Edifício
R, 1649-038 Lisboa, Portugal
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Marianna Katz
- Hovione
Farmaciência S.A., Estrada do Paço do Lumiar, Campus do Lumiar, Edifício
R, 1649-038 Lisboa, Portugal
| | - Maria M. M. Santos
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Carlos A. M. Afonso
- Research
Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
48
|
Homma K, Miura Y, Kobayashi M, Chintrakulchai W, Toyoda M, Ogi K, Michinishi J, Ohtake T, Honda Y, Nomoto T, Takemoto H, Nishiyama N. Fine tuning of the net charge alternation of polyzwitterion surfaced lipid nanoparticles to enhance cellular uptake and membrane fusion potential. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2338785. [PMID: 38646148 PMCID: PMC11028023 DOI: 10.1080/14686996.2024.2338785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/25/2024] [Indexed: 04/23/2024]
Abstract
Lipid nanoparticles (LNPs) coated with functional and biocompatible polymers have been widely used as carriers to deliver oligonucleotide and messenger RNA therapeutics to treat diseases. Poly(ethylene glycol) (PEG) is a representative material used for the surface coating, but the PEG surface-coated LNPs often have reduced cellular uptake efficiency and pharmacological activity. Here, we demonstrate the effect of pH-responsive ethylenediamine-based polycarboxybetaines with different molecular weights as an alternative structural component to PEG for the coating of LNPs. We found that appropriate tuning of the molecular weight around polycarboxybetaine-modified LNP, which incorporated small interfering RNA, could enhance the cellular uptake and membrane fusion potential in cancerous pH condition, thereby facilitating the gene silencing effect. This study demonstrates the importance of the design and molecular length of polymers on the LNP surface to provide effective drug delivery to cancer cells.
Collapse
Affiliation(s)
- Keitaro Homma
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Motoaki Kobayashi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Wanphiwat Chintrakulchai
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Masahiro Toyoda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Koichi Ogi
- I&S Department, Corporate R&D division, NOF CORPORATION, Kanagawa, Japan
| | - Junya Michinishi
- I&S Department, Corporate R&D division, NOF CORPORATION, Kanagawa, Japan
| | - Tomoyuki Ohtake
- I&S Department, Corporate R&D division, NOF CORPORATION, Kanagawa, Japan
| | - Yuto Honda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kanagawa, Japan
| | - Takahiro Nomoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyasu Takemoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kanagawa, Japan
| |
Collapse
|
49
|
López-Estévez AM, Lapuhs P, Pineiro-Alonso L, Alonso MJ. Personalized Cancer Nanomedicine: Overcoming Biological Barriers for Intracellular Delivery of Biopharmaceuticals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309355. [PMID: 38104275 DOI: 10.1002/adma.202309355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/09/2023] [Indexed: 12/19/2023]
Abstract
The success of personalized medicine in oncology relies on using highly effective and precise therapeutic modalities such as small interfering RNA (siRNA) and monoclonal antibodies (mAbs). Unfortunately, the clinical exploitation of these biological drugs has encountered obstacles in overcoming intricate biological barriers. Drug delivery technologies represent a plausible strategy to overcome such barriers, ultimately facilitating the access to intracellular domains. Here, an overview of the current landscape on how nanotechnology has dealt with protein corona phenomena as a first and determinant biological barrier is presented. This continues with the analysis of strategies facilitating access to the tumor, along with conceivable methods for enhanced tumor penetration. As a final step, the cellular barriers that nanocarriers must confront in order for their biological cargo to reach their target are deeply analyzed. This review concludes with a critical analysis and future perspectives of the translational advances in personalized oncological nanomedicine.
Collapse
Affiliation(s)
- Ana María López-Estévez
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Philipp Lapuhs
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Laura Pineiro-Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| |
Collapse
|
50
|
Geng X, Liang X, Liu Y, Chen Y, Xue B, Wei X, Yuan Z. Natural Fat Nanoemulsions for Enhanced Optical Coherence Tomography Neuroimaging and Tumor Imaging in the Second Near-Infrared Window. ACS NANO 2024; 18:9187-9198. [PMID: 38466960 DOI: 10.1021/acsnano.4c01204] [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: 03/13/2024]
Abstract
Optical coherence tomography (OCT) imaging mainly uses backscattered light to visualize the structural and functional information on biological tissues. In particular, OCT angiography can not only map the capillary networks but also capture the blood flow in the tissue microenvironment, making it a good candidate for neuroimaging and tumor imaging in vivo and in real time. To further improve the detection accuracy of cancer or brain disorders, it is essential to develop a natural and nontoxic contrast agent for enhanced OCT imaging in the second near-infrared (NIR-II) window. In this study, a superior biocompatible and highly scattering NIR-II fat nanoemulsion was constructed to improve OCT imaging contrast and depth for monitoring the vascular network changes of the cerebral cortex or tumor. In vivo experimental results demonstrated that a natural fat nanoemulsion can serve as an excellent probe for enhanced OCT neuroimaging and tumor imaging.
Collapse
Affiliation(s)
- Xiaorui Geng
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Xiao Liang
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR 999078, China
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yubin Liu
- College of Photonics and Electric Engineering, Fuzhou Normal University, Fuzhou, 350117, China
| | - Yuhao Chen
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Bin Xue
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR 999078, China
- Shenzhen Key Laboratory of Ultraintense Laser and Advanced Material Technology, Center for Advanced Material Diagnostic Technology, and College of Engineering Physics, Shenzhen Technology University, Shenzhen, 518118, China
| | - Xianyuan Wei
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR 999078, China
| | - Zhen Yuan
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR 999078, China
| |
Collapse
|