1
|
Alter CL, Lotter C, Puligilla RD, Bolten JS, Sedzicki J, Marchese J, Schittny V, Rucci F, Beverly M, Palivan CG, Detampel P, Einfalt T, Huwyler J. Nano Plasma Membrane Vesicle-Lipid Nanoparticle Hybrids for Enhanced Gene Delivery and Expression. Adv Healthc Mater 2025; 14:e2401888. [PMID: 39523736 DOI: 10.1002/adhm.202401888] [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: 05/24/2024] [Revised: 08/31/2024] [Indexed: 11/16/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as the leading nonviral nucleic acid (NA) delivery system, gaining widespread attention for their use in COVID-19 vaccines. They are recognized for their efficient NA encapsulation, modifiability, and scalable production. However, LNPs face efficacy and potency limitations due to suboptimal intracellular processing, with endosomal escape efficiencies (ESE) below 2.5%. Additionally, up to 70% of NPs undergo recycling and exocytosis after cellular uptake. In contrast, cell-derived vesicles offer biocompatibility and high-delivery efficacy but are challenging to load with exogenous NAs and to manufacture at large-scale. To leverage the strengths of both systems, a hybrid system is designed by combining cell-derived vesicles, such as nano plasma membrane vesicles (nPMVs), with LNPs through microfluidic mixing and subsequent dialysis. These hybrids demonstrate up to tenfold increase in ESE and an 18-fold rise in reporter gene expression in vitro and in vivo in zebrafish larvae (ZFL) and mice, compared to traditional LNPs. These improvements are linked to their unique physico-chemical properties, composition, and morphology. By incorporating cell-derived vesicles, this strategy streamlines the development process, significantly enhancing the efficacy and potency of gene delivery systems without the need for extensive screening.
Collapse
Affiliation(s)
- Claudio Luca Alter
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
- Swiss Nanoscience Institute, University of Basel, Klingelbergstrasse 82, Basel, 4056, Switzerland
| | - Claudia Lotter
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
| | - Ramya Deepthi Puligilla
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
| | - Jan Stephan Bolten
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
| | - Jaroslaw Sedzicki
- Biozentrum, University of Basel, Spitalstrasse 41, Basel, 4056, Switzerland
| | - Jason Marchese
- Novartis BioMedical Research, 100 Technology Square, Cambridge, MA, 02139, USA
| | - Valentin Schittny
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
| | - Francesca Rucci
- Novartis Biologics Research Center, Fabrikstrasse 16, Basel, 4056, Switzerland
| | - Michael Beverly
- Novartis BioMedical Research, 100 Technology Square, Cambridge, MA, 02139, USA
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, Basel, 4058, Switzerland
| | - Pascal Detampel
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
| | - Tomaž Einfalt
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
| | - Jörg Huwyler
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
| |
Collapse
|
2
|
AbouSamra MM. Liposomal nano-carriers mediated targeting of liver disorders: mechanisms and applications. J Liposome Res 2024; 34:728-743. [PMID: 38988127 DOI: 10.1080/08982104.2024.2377085] [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: 04/18/2024] [Revised: 06/12/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
Liver disorders present a significant global health challenge, necessitating the exploration of innovative treatment modalities. Liposomal nanocarriers have emerged as promising candidates for targeted drug delivery to the liver. This review offers a comprehensive examination of the mechanisms and applications of liposomal nanocarriers in addressing various liver disorders. Firstly discussing the liver disorders and the conventional treatment approaches, the review delves into the liposomal structure and composition. Moreover, it tackles the different mechanisms of liposomal targeting including both passive and active strategies. After that, the review moves on to explore the therapeutic potentials of liposomal nanocarriers in treating liver cirrhosis, fibrosis, viral hepatitis, and hepatocellular carcinoma. Through discussing recent advancements and envisioning future perspectives, this review highlights the role of liposomal nanocarriers in enhancing the effectiveness and the safety of liver disorders and consequently improving patient outcomes and enhances life quality.
Collapse
Affiliation(s)
- Mona M AbouSamra
- Pharmaceutical Technology Department, National Research Centre, Giza, Egypt
| |
Collapse
|
3
|
Cui J, Wen Z, Huang H, Qin S, Luo Y, Zhang W, Wu W. The Pharmacokinetics and Liver-Targeting Evaluation of Silybin Liposomes Mediated by the NTCP/OCTN2 Dual Receptors. Mol Pharm 2024; 21:4912-4923. [PMID: 39370820 DOI: 10.1021/acs.molpharmaceut.3c01245] [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: 10/08/2024]
Abstract
The disadvantage of a traditional dosage regimen is the inability to deliver a sufficient drug concentration to the lesion site, which can result in adverse side effects due to nonspecific drug delivery. Actively targeting hepatic cells is a promising therapeutic strategy for liver disease. In this study, l-carnitine and a targeting peptide derived from the hepatitis B virus large envelope protein were used to modify liposomes for drug delivery to the liver through the sodium taurocholate cotransporting polypeptide (NTCP) and the organic cation/carnitine transporter 2 (OCTN2) receptors. Silybin was selected as the model drug. The solubility of silybin can reach 0.3 mg/mL after encapsulation in liposomes. The NTCP-specific and OCTN2-accelerated Myrcludex B and l-carnitine dual-modified liposomes were validated in vitro. The uptake of coumarin-6 in dual ligand-modified liposomes by hepatocytes was up to 2.36 μg/mg compared with unmodified liposomes (1.05 μg/mg). The pharmacokinetics and targeting abilities of various liposome formulations were evaluated in Kunming mice. Targeted liposomes increased the concentration of silybin and prolonged the drug's retention time in the liver. The area under the liver's pharmacokinetic curve of targeted liposomes was twice that of silybin injection, suggesting the promising application potential of silybin-loaded hepatotropic nanovesicles.
Collapse
Affiliation(s)
- Jian Cui
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Huajie Huang
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Shuilin Qin
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Yanjie Luo
- Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi 541002, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi 541199, China
| |
Collapse
|
4
|
Unagolla JM, Das S, Flanagan R, Oehler M, Menon JU. Targeting chronic liver diseases: Molecular markers, drug delivery strategies and future perspectives. Int J Pharm 2024; 660:124381. [PMID: 38917958 PMCID: PMC11246230 DOI: 10.1016/j.ijpharm.2024.124381] [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: 01/28/2024] [Revised: 06/10/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
Chronic liver inflammation, a pervasive global health issue, results in millions of annual deaths due to its progression from fibrosis to the more severe forms of cirrhosis and hepatocellular carcinoma (HCC). This insidious condition stems from diverse factors such as obesity, genetic conditions, alcohol abuse, viral infections, autoimmune diseases, and toxic accumulation, manifesting as chronic liver diseases (CLDs) such as metabolic dysfunction-associated steatotic liver disease (MASLD), metabolic dysfunction-associated steatohepatitis (MASH), alcoholic liver disease (ALD), viral hepatitis, drug-induced liver injury, and autoimmune hepatitis. Late detection of CLDs necessitates effective treatments to inhibit and potentially reverse disease progression. However, current therapies exhibit limitations in consistency and safety. A potential breakthrough lies in nanoparticle-based drug delivery strategies, offering targeted delivery to specific liver cell types, such as hepatocytes, Kupffer cells, and hepatic stellate cells. This review explores molecular targets for CLD treatment, ongoing clinical trials, recent advances in nanoparticle-based drug delivery, and the future outlook of this research field. Early intervention is crucial for chronic liver disease. Having a comprehensive understanding of current treatments, molecular biomarkers and novel nanoparticle-based drug delivery strategies can have enormous impact in guiding future strategies for the prevention and treatment of CLDs.
Collapse
Affiliation(s)
- Janitha M Unagolla
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Subarna Das
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Riley Flanagan
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881, USA
| | - Marin Oehler
- Department of Biomedical Engineering, College of Engineering, University of Rhode Island, Kingston, RI 02881, USA
| | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA; Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881, USA.
| |
Collapse
|
5
|
Yuan Z, Yan R, Fu Z, Wu T, Ren C. Impact of physicochemical properties on biological effects of lipid nanoparticles: Are they completely safe. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172240. [PMID: 38582114 DOI: 10.1016/j.scitotenv.2024.172240] [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: 11/09/2023] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Lipid nanoparticles (LNPs) are promising materials and human-use approved excipients, with manifold applications in biomedicine. Researchers have tended to focus on improving the pharmacological efficiency and organ targeting of LNPs, while paid relatively less attention to the negative aspects created by their specific physicochemical properties. Here, we discuss the impacts of LNPs' physicochemical properties (size, surface hydrophobicity, surface charge, surface modification and lipid composition) on the adsorption-transportation-distribution-clearance processes and bio-nano interactions. In addition, since there is a lack of review emphasizing on toxicological profiles of LNPs, this review outlined immunogenicity, inflammation, hemolytic toxicity, cytotoxicity and genotoxicity induced by LNPs and the underlying mechanisms, with the aim to understand the properties that underlie the biological effects of these materials. This provides a basic strategy that increased efficacy of medical application with minimized side-effects can be achieved by modulating the physicochemical properties of LNPs. Therefore, addressing the effects of physicochemical properties on toxicity induced by LNPs is critical for understanding their environmental and health risks and will help clear the way for LNPs-based drugs to eventually fulfill their promise as a highly effective therapeutic agents for diverse diseases in clinic.
Collapse
Affiliation(s)
- Ziyi Yuan
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Ruyu Yan
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Zuyi Fu
- College of Rehabilitation, Captital Medical University, Beijing, China
| | - Tao Wu
- Beijing Key Laboratory of Enze Biomass Fine Chemicals, College of New Materials and Chemical Engineering, Beijing Institute of Petrochemical Technology, Beijing, China.
| | - Chaoxiu Ren
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Qin Y, Ou L, Zha L, Zeng Y, Li L. Delivery of nucleic acids using nanomaterials. MOLECULAR BIOMEDICINE 2023; 4:48. [PMID: 38092998 PMCID: PMC10719232 DOI: 10.1186/s43556-023-00160-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
The increasing number of approved nucleic acid therapeutics demonstrates the potential for the prevention and treatment of a broad spectrum of diseases. This trend underscores the significant impact and promise of nucleic acid-based treatments in the field of medicine. Nevertheless, employing nucleic acids as therapeutics is challenging due to their susceptibility to degradation by nucleases and their unfavorable physicochemical characteristics that hinder delivery into cells. Appropriate vectors play a pivotal role in improving nucleic acid stability and delivering nucleic acids into specific cells. The maturation of delivery systems has led to breakthroughs in the development of therapeutics based on nucleic acids such as DNA, siRNA, and mRNA. Non-viral vectors have gained prominence among the myriad of nanomaterials due to low immunogenicity, ease of manufacturing, and simplicity of cost-effective, large-scale production. Here, we provide an overview of the recent advancements in nanomaterials for nucleic acid delivery. Specifically, we give a detailed introduction to the characteristics of polymers, lipids, and polymer-lipid hybrids, and provide comprehensive descriptions of their applications in nucleic acid delivery. Also, biological barriers, administration routes, and strategies for organ-selective delivery of nucleic acids are discussed. In summary, this review offers insights into the rational design of next-generation delivery vectors for nucleic acid delivery.
Collapse
Affiliation(s)
- Yuyang Qin
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Liyuan Ou
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Lili Zha
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Yue Zeng
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Ling Li
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Yang C, Mwangi SM, Balasubramaniam A, Li G, Merlin-Zhang O, Liu Y, Srinivasan S. Treatment of Obesity Through Glial Cell-Derived Neurotrophic Factor Lipid Nanoparticle Delivery in Mice. GASTRO HEP ADVANCES 2023; 3:38-47. [PMID: 38390283 PMCID: PMC10883424 DOI: 10.1016/j.gastha.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
BACKGROUND AND AIMS The overexpression of glial cell-derived neurotrophic factor (GDNF) in the liver and adipose tissues offers strong protection against high-fat diet (HFD)-induced obesity in mice. We hypothesize that sustainably enhancing GDNF expression in the liver may provide a therapeutic effect that can prevent the progression of HFD-induced obesity in mice. METHODS Expression lentivector encoding mouse GDNF (GDNF(pDNA) or empty vector (pDNA, control) were encapsulated in lipid nanoparticles (LNPs) using the thin-film hydration method. Mice were fed with regular diet (RD) or HFD for 20 weeks prior to injection and the GDNF and control vector-loaded LNPs were administered by intravenous (IV) injection to mice once weekly for 5 weeks. Changes in body weight were monitored and mice tissues were collected and imaged for fluorescence using an IVIS in vivo imaging system. Post-treatment abdominal fat weight, colon length, and spleen weight were obtained. GDNF protein levels in the liver and serum were quantified by enzyme-linked immunosorbent assay, while liver AKT serine/threonine kinase and AMP-activated protein kinase phosphorylation levels were evaluated by Western blotting. RESULTS IV-injected GDNF(pDNA)-loaded LNPs targeted the liver and remained in there for up to 15 days postinjection. A single injection of GDNF(pDNA)-loaded LNPs significantly increased GDNF expression for 7 days and consequently increased the levels of phosphorylated AKT serine/threonine kinase and AMP-activated protein kinase. Once weekly injections of GDNF(pDNA)-loaded LNPs for 5 weeks slowed increase in body weight, reduced abdominal fat, and modulated the gut microbiota toward a healthier composition in HFD-fed mice. CONCLUSION GDNF(pDNA)-loaded LNPs could potentially be developed as a therapeutic strategy to reverse weight gain in obese patients.
Collapse
Affiliation(s)
- Chunhua Yang
- Institute for Biomedical Sciences, Center for Inflammation, Immunity & Infection, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
| | - Simon Musyoka Mwangi
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| | - Arun Balasubramaniam
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| | - Ge Li
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| | | | - Yunshan Liu
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| | - Shanthi Srinivasan
- Department of Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, Georgia
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
8
|
Abstract
Nanomedicine represents a promising way to devise better drug delivery systems (DDSs), and the development of cell/tissue-based lipid carriers is a promising strategy. In this study, the author proposes the concept of reconstituted lipid nanoparticles (rLNPs) and offers a facile preparation method. The results demonstrated that the preparation of ultrasmall (∼20 nm) rLNPs can be highly reproducible from both cells (a mouse breast cancer cell line, 4T1) and tissue (mouse liver tissue). As a selected model platform, rLNPs derived from mouse liver tissue can be further labeled with imaging molecules (indocyanine green and coumarin 6) and modified with targeting moiety (biotin). Moreover, rLNPs were proved to be highly biocompatible and able to load various drugs, such as doxorubicin hydrochloride (Dox) and curcumin (Cur). Most importantly, Dox-loaded rLNPs (rLNPs/Dox) exerted good in vitro and in vivo anticancer performances. Therefore, rLNPs might be a potential versatile carrier for the construction of different DDSs and treatment of a variety of diseases.
Collapse
Affiliation(s)
- Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, Jiangsu, P. R. China
| |
Collapse
|
9
|
Alter CL, Detampel P, Schefer RB, Lotter C, Hauswirth P, Puligilla RD, Weibel VJ, Schenk SH, Heusermann W, Schürz M, Meisner-Kober N, Palivan C, Einfalt T, Huwyler J. High efficiency preparation of monodisperse plasma membrane derived extracellular vesicles for therapeutic applications. Commun Biol 2023; 6:478. [PMID: 37137966 PMCID: PMC10156699 DOI: 10.1038/s42003-023-04859-2] [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: 02/18/2022] [Accepted: 04/21/2023] [Indexed: 05/05/2023] Open
Abstract
Extracellular vesicles (EVs) are highly interesting for the design of next-generation therapeutics. However, their preparation methods face challenges in standardization, yield, and reproducibility. Here, we describe a highly efficient and reproducible EV preparation method for monodisperse nano plasma membrane vesicles (nPMVs), which yields 10 to 100 times more particles per cell and hour than conventional EV preparation methods. nPMVs are produced by homogenizing giant plasma membrane vesicles following cell membrane blebbing and apoptotic body secretion induced by chemical stressors. nPMVs showed no significant differences compared to native EVs from the same cell line in cryo-TEM analysis, in vitro cellular interactions, and in vivo biodistribution studies in zebrafish larvae. Proteomics and lipidomics, on the other hand, suggested substantial differences consistent with the divergent origin of these two EV types and indicated that nPMVs primarily derive from apoptotic extracellular vesicles. nPMVs may provide an attractive source for developing EV-based pharmaceutical therapeutics.
Collapse
Affiliation(s)
- Claudio L Alter
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
- Swiss Nanoscience Institute, University of Basel, Klingelbergstrasse 82, 4056, Basel, Switzerland
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058, Basel, Switzerland
| | - Pascal Detampel
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Roman B Schefer
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Claudia Lotter
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Patrick Hauswirth
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Ramya D Puligilla
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Vera J Weibel
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Susanne H Schenk
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Wolf Heusermann
- Imaging Core Facility, University of Basel, Spitalstrasse 41, 4056, Basel, Switzerland
| | - Melanie Schürz
- Department of Biosciences & Medical Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
| | - Nicole Meisner-Kober
- Department of Biosciences & Medical Biology, University of Salzburg, Hellbrunnerstrasse 34, 5020, Salzburg, Austria
| | - Cornelia Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, BPR 1096, 4058, Basel, Switzerland
| | - Tomaž Einfalt
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Jörg Huwyler
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
10
|
Cheng D, Wen Z, Chen H, Lin S, Zhang W, Tang X, Wu W. Hepatocyte-targeting and tumor microenvironment-responsive liposomes for enhanced anti-hepatocarcinoma efficacy. Drug Deliv 2022; 29:2995-3008. [PMID: 36104946 PMCID: PMC9487930 DOI: 10.1080/10717544.2022.2122635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
To increase the antitumor drug concentration in the liver tumor site and improve the therapeutic effects, a functionalized liposome (PPP-LIP) with tumor targetability and enhanced internalization after matrix metalloproteinase-2 (MMP2)-triggered cell-penetrating peptide (TATp) exposure was modified with myrcludex B (a synthetic HBV preS-derived lipopeptide endowed with compelling liver tropism) for liver tumor-specific delivery. After intravenous administration, PPP-LIP was mediated by myrcludex B to reach the hepatocyte surface. The MMP2-overexpressing tumor microenvironment deprotected PEG, exposing it to TATp, facilitating tumor penetration and subsequent efficient destruction of tumor cells. In live imaging of small animals and cellular uptake, PPP-LIP was taken up much more than typical unmodified liposomes in the ICR mouse liver and liver tumor cells. Hydroxycamptothecin (HCPT)-loaded PPP-LIP showed a better antitumor effect than commercially available HCPT injections among MTT, three-dimensional (3 D) tumor ball, and tumor-bearing nude mouse experiments. Our findings indicated that PPP-LIP nanocarriers could be a promising tumor-targeted medication delivery strategy for treating liver cancers with elevated MMP2 expression.
Collapse
Affiliation(s)
- Dongliang Cheng
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Hui Chen
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Shiyuan Lin
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Xin Tang
- School of Public Health, Guilin Medical University, Guilin, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin, China
| |
Collapse
|
11
|
Ma XY, Hill BD, Hoang T, Wen F. Virus-inspired strategies for cancer therapy. Semin Cancer Biol 2022; 86:1143-1157. [PMID: 34182141 PMCID: PMC8710185 DOI: 10.1016/j.semcancer.2021.06.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 01/27/2023]
Abstract
The intentional use of viruses for cancer therapy dates back over a century. As viruses are inherently immunogenic and naturally optimized delivery vehicles, repurposing viruses for drug delivery, tumor antigen presentation, or selective replication in cancer cells represents a simple and elegant approach to cancer treatment. While early virotherapy was fraught with harsh side effects and low response rates, virus-based therapies have recently seen a resurgence due to newfound abilities to engineer and tune oncolytic viruses, virus-like particles, and virus-mimicking nanoparticles for improved safety and efficacy. However, despite their great potential, very few virus-based therapies have made it through clinical trials. In this review, we present an overview of virus-inspired approaches for cancer therapy, discuss engineering strategies to enhance their mechanisms of action, and highlight their application for overcoming the challenges of traditional cancer therapies.
Collapse
Affiliation(s)
- Xiao Yin Ma
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brett D Hill
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Trang Hoang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
12
|
Davis G, Kurse A, Agarwal A, Sheikh-Hamad D, Kumar MNVR. Nano-encapsulation strategies to circumvent drug-induced kidney injury and targeted nanomedicines to treat kidney diseases. CURRENT OPINION IN TOXICOLOGY 2022. [DOI: 10.1016/j.cotox.2022.100346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Li Y, Zhang R, Xu Z, Wang Z. Advances in Nanoliposomes for the Diagnosis and Treatment of Liver Cancer. Int J Nanomedicine 2022; 17:909-925. [PMID: 35250267 PMCID: PMC8893038 DOI: 10.2147/ijn.s349426] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
The mortality rate of liver cancer is gradually increasing worldwide due to the increasing risk factors such as fatty liver, diabetes, and alcoholic cirrhosis. The diagnostic methods of liver cancer include ultrasound (US), computed tomography (CT), and magnetic resonance imaging (MRI), among others. The treatment of liver cancer includes surgical resection, transplantation, ablation, and chemoembolization; however, treatment still faces multiple challenges due to its insidious development, high rate of recurrence after surgical resection, and high failure rate of transplantation. The emergence of liposomes has provided new insights into the treatment of liver cancer. Due to their excellent carrier properties and maneuverability, liposomes can be used to perform a variety of functions such as aiding in imaging diagnoses, combinatorial therapies, and integrating disease diagnosis and treatment. In this paper, we further discuss such advantages.
Collapse
Affiliation(s)
- Yitong Li
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, Jilin, People’s Republic of China
| | - Ruihang Zhang
- Second Clinical Medical College, Henan University of Traditional Chinese Medicine, Zhengzhou, 450052, Henan, People’s Republic of China
| | - Zhen Xu
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, Jilin, People’s Republic of China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, Changchun, 130021, Jilin, People’s Republic of China
- Correspondence: Zhicheng Wang, NHC Key Laboratory of Radiobiology (Jilin University), School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, 130021, Jilin, People’s Republic of China, Tel +86 13843131059, Fax +86 431185619443, Email
| |
Collapse
|
14
|
Impact of Linker Modification and PEGylation of Vancomycin Conjugates on Structure-Activity Relationships and Pharmacokinetics. Pharmaceuticals (Basel) 2022; 15:ph15020159. [PMID: 35215272 PMCID: PMC8880691 DOI: 10.3390/ph15020159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
As multidrug-resistant bacteria represent a concerning burden, experts insist on the need for a dramatic rethinking on antibiotic use and development in order to avoid a post-antibiotic era. New and rapidly developable strategies for antimicrobial substances, in particular substances highly potent against multidrug-resistant bacteria, are urgently required. Some of the treatment options currently available for multidrug-resistant bacteria are considerably limited by side effects and unfavorable pharmacokinetics. The glycopeptide vancomycin is considered an antibiotic of last resort. Its use is challenged by bacterial strains exhibiting various types of resistance. Therefore, in this study, highly active polycationic peptide-vancomycin conjugates with varying linker characteristics or the addition of PEG moieties were synthesized to optimize pharmacokinetics while retaining or even increasing antimicrobial activity in comparison to vancomycin. The antimicrobial activity of the novel conjugates was determined by microdilution assays on susceptible and vancomycin-resistant bacterial strains. VAN1 and VAN2, the most promising linker-modified derivatives, were further characterized in vivo with molecular imaging and biodistribution studies in rodents, showing that the linker moiety influences both antimicrobial activity and pharmacokinetics. Encouragingly, VAN2 was able to undercut the resistance breakpoint in microdilution assays on vanB and vanC vancomycin-resistant enterococci. Out of all PEGylated derivatives, VAN:PEG1 and VAN:PEG3 were able to overcome vanC resistance. Biodistribution studies of the novel derivatives revealed significant changes in pharmacokinetics when compared with vancomycin. In conclusion, linker modification of vancomycin-polycationic peptide conjugates represents a promising strategy for the modulation of pharmacokinetic behavior while providing potent antimicrobial activity.
Collapse
|
15
|
Shepherd SJ, Warzecha CC, Yadavali S, El-Mayta R, Alameh MG, Wang L, Weissman D, Wilson JM, Issadore D, Mitchell MJ. Scalable mRNA and siRNA Lipid Nanoparticle Production Using a Parallelized Microfluidic Device. NANO LETTERS 2021; 21:5671-5680. [PMID: 34189917 PMCID: PMC10726372 DOI: 10.1021/acs.nanolett.1c01353] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
A major challenge to advance lipid nanoparticles (LNPs) for RNA therapeutics is the development of formulations that can be produced reliably across the various scales of drug development. Microfluidics can generate LNPs with precisely defined properties, but have been limited by challenges in scaling throughput. To address this challenge, we present a scalable, parallelized microfluidic device (PMD) that incorporates an array of 128 mixing channels that operate simultaneously. The PMD achieves a >100× production rate compared to single microfluidic channels, without sacrificing desirable LNP physical properties and potency typical of microfluidic-generated LNPs. In mice, we show superior delivery of LNPs encapsulating either Factor VII siRNA or luciferase-encoding mRNA generated using a PMD compared to conventional mixing, with a 4-fold increase in hepatic gene silencing and 5-fold increase in luciferase expression, respectively. These results suggest that this PMD can generate scalable and reproducible LNP formulations needed for emerging clinical applications, including RNA therapeutics and vaccines.
Collapse
Affiliation(s)
- Sarah J. Shepherd
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Claude C. Warzecha
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sagar Yadavali
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Mohamad-Gabriel Alameh
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Lili Wang
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - James M. Wilson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - David Issadore
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States; Department of Electrical and Systems Engineering and Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States; Abramson Cancer Center, Perelman School of Medicine, Institute for Immunology, Perelman School of Medicine, Cardiovascular Institute, Perelman School of Medicine, and Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
16
|
Liu M, Fang X, Yang Y, Wang C. Peptide-Enabled Targeted Delivery Systems for Therapeutic Applications. Front Bioeng Biotechnol 2021; 9:701504. [PMID: 34277592 PMCID: PMC8281044 DOI: 10.3389/fbioe.2021.701504] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Receptor-targeting peptides have been extensively pursued for improving binding specificity and effective accumulation of drugs at the site of interest, and have remained challenging for extensive research efforts relating to chemotherapy in cancer treatments. By chemically linking a ligand of interest to drug-loaded nanocarriers, active targeting systems could be constructed. Peptide-functionalized nanostructures have been extensively pursued for biomedical applications, including drug delivery, biological imaging, liquid biopsy, and targeted therapies, and widely recognized as candidates of novel therapeutics due to their high specificity, well biocompatibility, and easy availability. We will endeavor to review a variety of strategies that have been demonstrated for improving receptor-specificity of the drug-loaded nanoscale structures using peptide ligands targeting tumor-related receptors. The effort could illustrate that the synergism of nano-sized structures with receptor-targeting peptides could lead to enrichment of biofunctions of nanostructures.
Collapse
Affiliation(s)
- Mingpeng Liu
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Department of Chemistry, Tsinghua University, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaocui Fang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanlian Yang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chen Wang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
17
|
Chander N, Morstein J, Bolten JS, Shemet A, Cullis PR, Trauner D, Witzigmann D. Optimized Photoactivatable Lipid Nanoparticles Enable Red Light Triggered Drug Release. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2008198. [PMID: 33880882 DOI: 10.1002/smll.202008198] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/25/2021] [Indexed: 06/12/2023]
Abstract
Encapsulation of small molecule drugs in long-circulating lipid nanoparticles (LNPs) can reduce toxic side effects and enhance accumulation at tumor sites. A fundamental problem, however, is the slow release of encapsulated drugs from these liposomal systems at the disease site resulting in limited therapeutic benefit. Methods to trigger release at specific sites are highly warranted. Here, it is demonstrated that incorporation of ultraviolet (UV-A) or red-light photoswitchable-phosphatidylcholine analogs (AzoPC and redAzoPC) in conventional LNPs generates photoactivatable LNPs (paLNPs) having comparable structural integrity, drug loading capacity, and size distribution to the parent DSPC-cholesterol liposomes. It is shown that 65-70% drug release (doxorubicin) can be induced from these systems by irradiation with pulsed light based on trans-to-cis azobenzene isomerization. In vitro it is confirmed that paLNPs are non-toxic in the dark but convey cytotoxicity upon irradiation in a human cancer cell line. In vivo studies in zebrafish embryos demonstrate prolonged blood circulation and extravasation of paLNPs comparable to clinically approved formulations, with enhanced drug release following irradiation with pulsed light. Conclusively, paLNPs closely mimic the properties of clinically approved LNPs with the added benefit of light-induced drug release making them promising candidates for clinical development.
Collapse
Affiliation(s)
- Nisha Chander
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Johannes Morstein
- Department of Chemistry, New York University, 100 Washington Square East, Room 712, New York, NY, 10003, USA
| | - Jan S Bolten
- Department of Pharmaceutical Technology, University of Basel, Klingelbergstrasse 50, Basel, 4056, Switzerland
| | - Andrej Shemet
- Department of Chemistry, New York University, 100 Washington Square East, Room 712, New York, NY, 10003, USA
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- NanoMedicines Innovation Network (NMIN), University of British Columbia, 2350 Health Sciences Mall, Room 5451, Vancouver, BC, V6T 1Z3, Canada
| | - Dirk Trauner
- Department of Chemistry, New York University, 100 Washington Square East, Room 712, New York, NY, 10003, USA
| | - Dominik Witzigmann
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- NanoMedicines Innovation Network (NMIN), University of British Columbia, 2350 Health Sciences Mall, Room 5451, Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
18
|
Pratsinis A, Uhl P, Bolten JS, Hauswirth P, Schenk SH, Urban S, Mier W, Witzigmann D, Huwyler J. Virus-Derived Peptides for Hepatic Enzyme Delivery. Mol Pharm 2021; 18:2004-2014. [PMID: 33844553 DOI: 10.1021/acs.molpharmaceut.0c01222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Recently, a lipopeptide derived from the hepatitis B virus (HBV) large surface protein has been developed as an HBV entry inhibitor. This lipopeptide, called MyrcludexB (MyrB), selectively binds to the sodium taurocholate cotransporting polypeptide (NTCP) on the basolateral membrane of hepatocytes. Here, the feasibility of coupling therapeutic enzymes to MyrB was investigated for the development of enzyme delivery strategies. Hepatotropic targeting shall enable enzyme prodrug therapies and detoxification procedures. Here, horseradish peroxidase (HRP) was conjugated to MyrB via maleimide chemistry, and coupling was validated by SDS-PAGE and reversed-phase HPLC. The specificity of the target recognition of HRP-MyrB could be shown in an NTCP-overexpressing liver parenchymal cell line, as demonstrated by competitive inhibition with an excess of free MyrB and displayed a strong linear dependency on the applied HRP-MyrB concentration. In vivo studies in zebrafish embryos revealed a dominating interaction of HRP-MyrB with scavenger endothelial cells vs xenografted NTCP expressing mammalian cells. In mice, radiolabeled 125I-HRP-MyrBy, as well as the non-NTCP targeted control HRP-peptide-construct (125I-HRP-alaMyrBy) demonstrated a strong liver accumulation confirming the nonspecific interaction with scavenger cells. Still, MyrB conjugation to HRP resulted in an increased and NTCP-mediated hepatotropism, as revealed by competitive inhibition. In conclusion, the model enzyme HRP was successfully conjugated to MyrB to achieve NTCP-specific targeting in vitro with the potential for ex vivo diagnostic applications. In vivo, target specificity was reduced by non-NTCP-mediated interactions. Nonetheless, tissue distribution experiments in zebrafish embryos provide mechanistic insight into underlying scavenging processes indicating partial involvement of stabilin receptors.
Collapse
Affiliation(s)
- Anna Pratsinis
- Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Philipp Uhl
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Jan Stephan Bolten
- Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Patrick Hauswirth
- Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Susanne Heidi Schenk
- Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Stephan Urban
- Department of Infectious Diseases, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Walter Mier
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Dominik Witzigmann
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.,NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, British ColumbiaV6T 1Z3, Canada
| | - Jörg Huwyler
- Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| |
Collapse
|
19
|
Loo YS, Bose RJ, McCarthy JR, Mat Azmi ID, Madheswaran T. Biomimetic bacterial and viral-based nanovesicles for drug delivery, theranostics, and vaccine applications. Drug Discov Today 2020; 26:902-915. [PMID: 33383213 DOI: 10.1016/j.drudis.2020.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/16/2020] [Accepted: 12/21/2020] [Indexed: 01/04/2023]
Abstract
Smart nanocarriers obtained from bacteria and viruses offer excellent biomimetic properties which has led to significant research into the creation of advanced biomimetic materials. Their versatile biomimicry has application as biosensors, biomedical scaffolds, immobilization, diagnostics, and targeted or personalized treatments. The inherent natural traits of biomimetic and bioinspired bacteria- and virus-derived nanovesicles show potential for their use in clinical vaccines and novel therapeutic drug delivery systems. The past few decades have seen significant progress in the bioengineering of bacteria and viruses to manipulate and enhance their therapeutic benefits. From a pharmaceutical perspective, biomimetics enable the safe integration of naturally occurring bacteria and virus particles to achieve high, stable rates of cellular transfection/infection and prolonged circulation times. In addition, biomimetic technologies can overcome safety concerns associated with live-attenuated and inactivated whole bacteria or viruses. In this review, we provide an update on the utilization of bacterial and viral particles as drug delivery systems, theranostic carriers, and vaccine/immunomodulation modalities.
Collapse
Affiliation(s)
- Yan Shan Loo
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia
| | - Rajendran Jc Bose
- Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY 13501, USA
| | - Jason R McCarthy
- Masonic Medical Research Institute, 2150 Bleecker St, Utica, NY 13501, USA
| | - Intan Diana Mat Azmi
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia.
| | - Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, International Medical University, No. 126 Jalan Jalil Perkasa 19, Bukit Jalil, 57000 Kuala Lumpur, Malaysia.
| |
Collapse
|
20
|
Kou L, Yao Q, Zhang H, Chu M, Bhutia YD, Chen R, Ganapathy V. Transporter-Targeted Nano-Sized Vehicles for Enhanced and Site-Specific Drug Delivery. Cancers (Basel) 2020; 12:E2837. [PMID: 33019627 PMCID: PMC7599460 DOI: 10.3390/cancers12102837] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Nano-devices are recognized as increasingly attractive to deliver therapeutics to target cells. The specificity of this approach can be improved by modifying the surface of the delivery vehicles such that they are recognized by the target cells. In the past, cell-surface receptors were exploited for this purpose, but plasma membrane transporters also hold similar potential. Selective transporters are often highly expressed in biological barriers (e.g., intestinal barrier, blood-brain barrier, and blood-retinal barrier) in a site-specific manner, and play a key role in the vectorial transfer of nutrients. Similarly, selective transporters are also overexpressed in the plasma membrane of specific cell types under pathological states to meet the biological needs demanded by such conditions. Nano-drug delivery systems could be strategically modified to make them recognizable by these transporters to enhance the transfer of drugs across the biological barriers or to selectively expose specific cell types to therapeutic drugs. Here, we provide a comprehensive review and detailed evaluation of the recent advances in the field of transporter-targeted nano-drug delivery systems. We specifically focus on areas related to intestinal absorption, transfer across blood-brain barrier, tumor-cell selective targeting, ocular drug delivery, identification of the transporters appropriate for this purpose, and details of the rationale for the approach.
Collapse
Affiliation(s)
- Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China;
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
- Department of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang 325035, China
| | - Hailin Zhang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
- Department of Children’s Respiration Disease, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Maoping Chu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China
| | - Yangzom D. Bhutia
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China;
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
| | - Vadivel Ganapathy
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang 325027, China;
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Zhejiang 325027, China; (Q.Y.); (H.Z.); (M.C.)
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| |
Collapse
|
21
|
Buck J, Mueller D, Mettal U, Ackermann M, Grisch-Chan HM, Thöny B, Zumbuehl A, Huwyler J, Witzigmann D. Improvement of DNA Vector Delivery of DOTAP Lipoplexes by Short-Chain Aminolipids. ACS OMEGA 2020; 5:24724-24732. [PMID: 33015490 PMCID: PMC7528285 DOI: 10.1021/acsomega.0c03303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/21/2020] [Indexed: 06/11/2023]
Abstract
Cellular delivery of DNA vectors for the expression of therapeutic proteins is a promising approach to treat monogenic disorders or cancer. Significant efforts in a preclinical and clinical setting have been made to develop potent nonviral gene delivery systems based on lipoplexes composed of permanently cationic lipids. However, transfection efficiency and tolerability of such systems are in most cases not satisfactory. Here, we present a one-pot combinatorial method based on double-reductive amination for the synthesis of short-chain aminolipids. These lipids can be used to maximize the DNA vector delivery when combined with the cationic lipid 1,2-dioleoyl-3-trimethylammonium propane (DOTAP). We incorporated various aminolipids into such lipoplexes to complex minicircle DNA and screened these systems in a human liver-derived cell line (HuH7) for gene expression and cytotoxicity. The lead aminolipid AL-A12 showed twofold enhanced gene delivery and reduced toxicity compared to the native DOTAP:cholesterol lipoplexes. Moreover, AL-A12-containing lipoplexes enabled enhanced transgene expression in vivo in the zebrafish embryo model.
Collapse
Affiliation(s)
- Jonas Buck
- Division
of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Dennis Mueller
- Department
of Chemistry, University of Fribourg, 1700 Fribourg, Switzerland
| | - Ute Mettal
- Department
of Chemistry, University of Fribourg, 1700 Fribourg, Switzerland
- Department
of Bioresources of the Fraunhofer Institute for Molecular Biology
and Applied Ecology, Institute for Insect
Biotechnology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Miriam Ackermann
- Department
of Chemistry, University of Fribourg, 1700 Fribourg, Switzerland
| | - Hiu Man Grisch-Chan
- Division
of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, 8032 Zürich, Switzerland
| | - Beat Thöny
- Division
of Metabolism and Children’s Research Center, University Children’s Hospital Zurich, 8032 Zürich, Switzerland
| | - Andreas Zumbuehl
- Department
of Chemistry, University of Fribourg, 1700 Fribourg, Switzerland
- Acthera
Therapeutics Ltd., Peter
Merian-Strasse 45, 4052 Basel, Switzerland
| | - Jörg Huwyler
- Division
of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Dominik Witzigmann
- Division
of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
22
|
Francia V, Schiffelers RM, Cullis PR, Witzigmann D. The Biomolecular Corona of Lipid Nanoparticles for Gene Therapy. Bioconjug Chem 2020; 31:2046-2059. [PMID: 32786370 DOI: 10.1021/acs.bioconjchem.0c00366] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Gene therapy holds great potential for treating almost any disease by gene silencing, protein expression, or gene correction. To efficiently deliver the nucleic acid payload to its target tissue, the genetic material needs to be combined with a delivery platform. Lipid nanoparticles (LNPs) have proven to be excellent delivery vectors for gene therapy and are increasingly entering into routine clinical practice. Over the past two decades, the optimization of LNP formulations for nucleic acid delivery has led to a well-established body of knowledge culminating in the first-ever RNA interference therapeutic using LNP technology, i.e., Onpattro, and many more in clinical development to deliver various nucleic acid payloads. Screening a lipid library in vivo for optimal gene silencing potency in hepatocytes resulted in the identification of the Onpattro formulation. Subsequent studies discovered that the key to Onpattro's liver tropism is its ability to form a specific "biomolecular corona". In fact, apolipoprotein E (ApoE), among other proteins, adsorbed to the LNP surface enables specific hepatocyte targeting. This proof-of-principle example demonstrates the use of the biomolecular corona for targeting specific receptors and cells, thereby opening up the road to rationally designing LNPs. To date, however, only a few studies have explored in detail the corona of LNPs, and how to efficiently modulate the corona remains poorly understood. In this review, we summarize recent discoveries about the biomolecular corona, expanding the knowledge gained with other nanoparticles to LNPs for nucleic acid delivery. In particular, we address how particle stability, biodistribution, and targeting of LNPs can be influenced by the biological environment. Onpattro is used as a case study to describe both the successful development of an LNP formulation for gene therapy and the key influence of the biological environment. Moreover, we outline the techniques available to isolate and analyze the corona of LNPs, and we highlight their advantages and drawbacks. Finally, we discuss possible implications of the biomolecular corona for LNP delivery and we examine the potential of exploiting the corona as a targeting strategy beyond the liver to develop next-generation gene therapies.
Collapse
Affiliation(s)
- Valentina Francia
- Department of Biochemistry and Molecular Biology, University of British Columbia, V6T 1Z3, Vancouver, British Columbia, Canada.,Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, 3584 CX, Utrecht, Netherlands
| | - Raymond M Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, 3584 CX, Utrecht, Netherlands
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, V6T 1Z3, Vancouver, British Columbia, Canada.,NanoMedicines Innovation Network (NMIN), University of British Columbia, V6T 1Z3, Vancouver, British Columbia, Canada
| | - Dominik Witzigmann
- Department of Biochemistry and Molecular Biology, University of British Columbia, V6T 1Z3, Vancouver, British Columbia, Canada.,NanoMedicines Innovation Network (NMIN), University of British Columbia, V6T 1Z3, Vancouver, British Columbia, Canada
| |
Collapse
|
23
|
Arias-Alpizar G, Kong L, Vlieg RC, Rabe A, Papadopoulou P, Meijer MS, Bonnet S, Vogel S, van Noort J, Kros A, Campbell F. Light-triggered switching of liposome surface charge directs delivery of membrane impermeable payloads in vivo. Nat Commun 2020; 11:3638. [PMID: 32686667 PMCID: PMC7371701 DOI: 10.1038/s41467-020-17360-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/25/2020] [Indexed: 01/14/2023] Open
Abstract
Surface charge plays a fundamental role in determining the fate of a nanoparticle, and any encapsulated contents, in vivo. Herein, we describe, and visualise in real time, light-triggered switching of liposome surface charge, from neutral to cationic, in situ and in vivo (embryonic zebrafish). Prior to light activation, intravenously administered liposomes, composed of just two lipid reagents, freely circulate and successfully evade innate immune cells present in the fish. Upon in situ irradiation and surface charge switching, however, liposomes rapidly adsorb to, and are taken up by, endothelial cells and/or are phagocytosed by blood resident macrophages. Coupling complete external control of nanoparticle targeting together with the intracellular delivery of encapsulated (and membrane impermeable) cargos, these compositionally simple liposomes are proof that advanced nanoparticle function in vivo does not require increased design complexity but rather a thorough understanding of the fundamental nano-bio interactions involved.
Collapse
Affiliation(s)
- Gabriela Arias-Alpizar
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Li Kong
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
- Tongji School of Pharmacy, Huazhong University of Science and Technology, 430030, Wuhan, P.R. China
| | - Redmar C Vlieg
- Leiden Institute of Physics (LION), Leiden University, P.O. Box 9504, 2300, RA, Leiden, The Netherlands
| | - Alexander Rabe
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230, Odense, Denmark
- BioNTech RNA Pharmaceuticals GmbH, An der Goldgrube 12, 55131, Mainz, Germany
| | - Panagiota Papadopoulou
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Michael S Meijer
- Department of Metals in Catalysis, Biomimetics & Inorganic Materials (MCBIM), Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Sylvestre Bonnet
- Department of Metals in Catalysis, Biomimetics & Inorganic Materials (MCBIM), Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Stefan Vogel
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230, Odense, Denmark
| | - John van Noort
- Leiden Institute of Physics (LION), Leiden University, P.O. Box 9504, 2300, RA, Leiden, The Netherlands
| | - Alexander Kros
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands.
| | - Frederick Campbell
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands.
| |
Collapse
|
24
|
Sedighi M, Rahimi F, Shahbazi MA, Rezayan AH, Kettiger H, Einfalt T, Huwyler J, Witzigmann D. Controlled Tyrosine Kinase Inhibitor Delivery to Liver Cancer Cells by Gate-Capped Mesoporous Silica Nanoparticles. ACS APPLIED BIO MATERIALS 2020; 3:239-251. [PMID: 35019440 DOI: 10.1021/acsabm.9b00772] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma is the most common type of primary malignancy in the liver and one of the most common types of cancer worldwide. Its readily increasing mortality rate highlights the urgent need for the development of efficient therapeutic strategies. Tyrosine kinase inhibitors (TKIs) such as sorafenib and sunitinib are used as efficient angiogenesis inhibitors for this purpose. However, despite their pharmacological effects, their transfer into clinical practice is characterized by their poor aqueous solubility and accumulation in off-target tissues, resulting in unfavorable side effects. Here, we report a nanocomposite made of amine-functionalized mesoporous silica nanocomposites (MSNs) that are surface-coated with cerium oxide nanoparticles (CNPs) for the controlled delivery and release of TKIs. Amine-functionalized MSNs were prepared using a sol-gel method and loaded with TKIs. To trap drug molecules into the mesoporous structure, CNPs were covalently conjugated to the surface of MSNs. The synthesis and functionalization steps were controlled using different characterization methods, confirming the desired morphology and structure, the identity of functional groups on the surface, successful coating, and appropriate loading efficiency. Under physiological conditions, CNP-capped MSNs demonstrated a sustained drug release over time as a result of CNPs' gatekeeping effect on the payloads. Strong cellular interactions with different liver cancer cells and enhanced cellular uptake were also observed in vitro for the gate-capped MSNs. Internalization of nanocomposites induced cell death via the production of reactive oxygen species, and subsequent activation of apoptosis pathways. This study demonstrates that gate-capped MSNs are promising chemotherapeutic vehicles characterized by a sustained drug release profile and high cellular internalization.
Collapse
Affiliation(s)
- Mahsa Sedighi
- Division of Nanobiotechnology, Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, 1439957131 Tehran, Iran.,Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056 Basel, Switzerland
| | - Fereshteh Rahimi
- Division of Nanobiotechnology, Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, 1439957131 Tehran, Iran
| | - Mohammad-Ali Shahbazi
- Department of Micro- and Nanotechnology, Technical University of Denmark, Ørsteds Plads, DK-2800 Kgs. Lyngby, Denmark.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 4513956184 Zanjan, Iran
| | - Ali Hossein Rezayan
- Division of Nanobiotechnology, Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, 1439957131 Tehran, Iran
| | - Helene Kettiger
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Tykistökatu 6A, FI-20520 Turku, Finland
| | - Tomaz Einfalt
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056 Basel, Switzerland
| | - Jörg Huwyler
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056 Basel, Switzerland
| | - Dominik Witzigmann
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, 4056 Basel, Switzerland.,Department of Biochemistry and Molecular Biology, University of British Columbia, Health Sciences Mall, V6T 1Z3 Vancouver, British Columbia, Canada
| |
Collapse
|