1
|
Li Y, Feng Q, Wang L, Gao X, Xi Y, Ye L, Ji J, Yang X, Zhai G. Current targeting strategies and advanced nanoplatforms for atherosclerosis therapy. J Drug Target 2024; 32:128-147. [PMID: 38217526 DOI: 10.1080/1061186x.2023.2300694] [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: 09/16/2023] [Accepted: 12/24/2023] [Indexed: 01/15/2024]
Abstract
Atherosclerosis is one of the major causes of death worldwide, and it is closely related to many cardiovascular diseases, such as stroke, myocardial infraction and angina. Although traditional surgical and pharmacological interventions can effectively retard or slow down the progression of atherosclerosis, it is very difficult to prevent or even reverse this disease. In recent years, with the rapid development of nanotechnology, various nanoagents have been designed and applied to different diseases including atherosclerosis. The unique atherosclerotic microenvironment with signature biological components allows nanoplatforms to distinguish atherosclerotic lesions from normal tissue and to approach plaques specifically. Based on the process of atherosclerotic plaque formation, this review summarises the nanodrug delivery strategies for atherosclerotic therapy, trying to provide help for researchers to understand the existing atherosclerosis management approaches as well as challenges and to reasonably design anti-atherosclerotic nanoplatforms.
Collapse
Affiliation(s)
- Yingchao Li
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Qixiang Feng
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Luyue Wang
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Xi Gao
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Yanwei Xi
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Lei Ye
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Jianbo Ji
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaoye Yang
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| | - Guangxi Zhai
- Department of Pharmaceutics, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
2
|
Tang X, Ding S, Yang S, Cheng Y, Liu H, Chen K, Han X. Polysorbate 80-containing ionizable lipid nanoparticles for mRNA delivery. Biomater Sci 2024; 12:5573-5581. [PMID: 39297400 DOI: 10.1039/d4bm00523f] [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: 10/23/2024]
Abstract
Ionizable lipid nanoparticles have demonstrated remarkable success as mRNA vaccine carriers and represent one of the most promising gene drug delivery vehicles. However, polyethylene glycol (PEG), one of the major components, can cause immunogenic reactions, anaphylaxis and increased blood clearance, leading to toxic side effects and reduced efficacy. In this study, we utilize polysorbate 80 (PS80) as a PEG alternative in formulating eGFP mRNA-loaded ionizable lipid nanoparticles (PS80-iLNPs), aiming to enhance stealth properties, uptake efficiency, and biosafety. Our findings revealed that PS80-iLNPs enhanced the stealthiness and resistance to serum interference. Compared to PEG-containing ionizable lipid nanoparticles (PEG-iLNPs), PS80-iLNPs showed a 1.14-fold increase in stealthiness. Moreover, at a total lipid concentration of 50 μg mL-1, PS80-iLNPs exhibited 1.12 times higher cell viability compared to PEG-iLNPs. Notably, under serum interference, PEG-iLNPs showed a 44.97% uptake reduction, whereas PS80-iLNPs exhibited a modest 30.55% decrease, underscoring its superior serum resistance. This work demonstrated that PS80 could serve as a suitable substitute for PEG, thus signifying an excellent basis for the development of PEG-free ionizable lipid nanoparticles.
Collapse
Affiliation(s)
- Xuefeng Tang
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Shixiao Ding
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Shilin Yang
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Yuqiao Cheng
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Hanyu Liu
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Kexin Chen
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Xiaojun Han
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
3
|
Yokoyama S, Kakeshita K, Imamura T, Shima T, Fujioka H, Yamazaki H, Koike T, Kinugawa K. Pegylated-liposomal Doxorubicin-induced Glomerular Thrombotic Microangiopathy. Intern Med 2024; 63:2839-2845. [PMID: 38462521 DOI: 10.2169/internalmedicine.3113-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Pegylated liposomal doxorubicin (PLD) has emerged as a recent innovation within the realm of antineoplastic agents, distinguished by its incorporation of doxorubicin within the liposomal bilayer. Given the low risk of cardiotoxicity, the clinical use of PLD has been expanding. We encountered a patient who underwent extended PLD therapy for recurrent malignancy and subsequently developed PLD-associated thrombotic microangiopathy, which was diagnosed by a detailed pathophysiological assessment. This case underscores the importance of considering thrombotic microangiopathy as a potential differential diagnosis in patients presenting with unexplained hypertension and renal impairment during prolonged PLD monotherapy.
Collapse
Affiliation(s)
- Shingo Yokoyama
- The Second Department of Internal Medicine, University of Toyama, Japan
| | - Kota Kakeshita
- The Second Department of Internal Medicine, University of Toyama, Japan
| | - Teruhiko Imamura
- The Second Department of Internal Medicine, University of Toyama, Japan
| | - Tomoko Shima
- Department of Obstetrics and Gynecology, University of Toyama, Japan
| | - Hayato Fujioka
- The Second Department of Internal Medicine, University of Toyama, Japan
| | - Hidenori Yamazaki
- The Second Department of Internal Medicine, University of Toyama, Japan
| | - Tsutomu Koike
- The Second Department of Internal Medicine, University of Toyama, Japan
| | - Koichiro Kinugawa
- The Second Department of Internal Medicine, University of Toyama, Japan
| |
Collapse
|
4
|
Shishlyannikov SM, Zubkov IN, Vysochinskaya VV, Gavrilova NV, Dobrovolskaya OA, Elpaeva EA, Maslov MA, Vasin A. Stable Polymer-Lipid Hybrid Nanoparticles Based on mcl-Polyhydroxyalkanoate and Cationic Liposomes for mRNA Delivery. Pharmaceutics 2024; 16:1305. [PMID: 39458633 PMCID: PMC11511049 DOI: 10.3390/pharmaceutics16101305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The development of polymer-lipid hybrid nanoparticles (PLNs) is a promising area of research, as it can help increase the stability of cationic lipid carriers. Hybrid PLNs are core-shell nanoparticle structures that combine the advantages of both polymer nanoparticles and liposomes, especially in terms of their physical stability and biocompatibility. Natural polymers such as polyhydroxyalkanoate (PHA) can be used as a matrix for the PLNs' preparation. Methods: In this study, we first obtained stable cationic hybrid PLNs using a cationic liposome (CL) composed of a polycationic lipid 2X3 (1,26-bis(cholest-5-en-3β-yloxycarbonylamino)-7,11,16,20-tetraazahexacosane tetrahydrochloride), helper lipid DOPE (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine), and the hydrophobic polymer mcl-PHA, which was produced by the soil bacterium Pseudomonas helmantisensis P1. Results: The new polymer-lipid carriers effectively encapsulated and delivered model mRNA-eGFP (enhanced green fluorescent protein mRNA) to BHK-21 cells. We then evaluated the role of mcl-PHA in increasing the stability of cationic PLNs in ionic solutions using dynamic light scattering data, electrophoretic mobility, and transmission electron microscopy techniques. Conclusions: The results showed that increasing the concentration of PBS (phosphate buffered saline) led to a decrease in the stability of the CLs. At high concentrations of PBS, the CLs aggregate. In contrast, the presence of isotonic PBS did not result in the aggregation of PLNs, and the particles remained stable for 120 h when stored at +4 °C. The obtained results show that PLNs hold promise for further in vivo studies on nucleic acid delivery.
Collapse
Affiliation(s)
- Sergey M. Shishlyannikov
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Politechnicheskaya St., 195251 Saint Petersburg, Russia; (I.N.Z.); (V.V.V.); (N.V.G.); (A.V.)
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popova Street, 197022 Saint Petersburg, Russia; (O.A.D.); (E.A.E.)
| | - Ilya N. Zubkov
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Politechnicheskaya St., 195251 Saint Petersburg, Russia; (I.N.Z.); (V.V.V.); (N.V.G.); (A.V.)
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popova Street, 197022 Saint Petersburg, Russia; (O.A.D.); (E.A.E.)
| | - Vera V. Vysochinskaya
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Politechnicheskaya St., 195251 Saint Petersburg, Russia; (I.N.Z.); (V.V.V.); (N.V.G.); (A.V.)
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popova Street, 197022 Saint Petersburg, Russia; (O.A.D.); (E.A.E.)
| | - Nina V. Gavrilova
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Politechnicheskaya St., 195251 Saint Petersburg, Russia; (I.N.Z.); (V.V.V.); (N.V.G.); (A.V.)
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popova Street, 197022 Saint Petersburg, Russia; (O.A.D.); (E.A.E.)
| | - Olga A. Dobrovolskaya
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popova Street, 197022 Saint Petersburg, Russia; (O.A.D.); (E.A.E.)
| | - Ekaterina A. Elpaeva
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popova Street, 197022 Saint Petersburg, Russia; (O.A.D.); (E.A.E.)
| | - Mikhail A. Maslov
- M.V. Lomonosov Institute of Fine Chemical Technologies, Rtu Mirea, 86 Vernadsky Ave., 119454 Moscow, Russia;
| | - Andrey Vasin
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Politechnicheskaya St., 195251 Saint Petersburg, Russia; (I.N.Z.); (V.V.V.); (N.V.G.); (A.V.)
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popova Street, 197022 Saint Petersburg, Russia; (O.A.D.); (E.A.E.)
| |
Collapse
|
5
|
Simon L, Reichel LS, Benkhaled BT, Devoisselle JM, Catrouillet S, Eberhardt J, Hoeppener S, Schubert US, Brendel JC, Morille M, Lapinte V, Traeger A. Polyoxazolines with Cholesterol Lipid Anchor for Fast Intracellular Delivery. Macromol Biosci 2024:e2400148. [PMID: 39374348 DOI: 10.1002/mabi.202400148] [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: 03/28/2024] [Revised: 08/01/2024] [Indexed: 10/09/2024]
Abstract
Due to the increasing challenges posed by the growing immunity to poly(ethylene glycol) (PEG), there is growing interest in innovative polymer-based materials as viable alternatives. In this study, the advantages of lipids and polymers are combined to allow efficient and rapid cytoplasmic drug delivery. Specifically, poly(2-methyl-2-oxazoline) is modified with a cholesteryl hemisuccinate group as a lipid anchor (CHEMSPOx). The CHEMSPOx is additionally functionalized with a coumarin group (CHEMSPOx-coumarin). Both polymers self-assembled in water into vesicles of ≈100 nm and are successfully loaded with a hydrophobic model drug. The loaded vesicles reveal high cellular internalization across variant cell lines within 1 h at 37 °C as well as 4 °C, albeit to a lesser extent. A kinetic study confirms the fast internalization within 5 min after the sample's addition. Therefore, different internalization pathways are involved, e.g., active uptake but also nonenergy dependent mechanisms. CHEMSPOx and CHEMSPOx-coumarin further demonstrate excellent cyto-, hemo-, and membrane compatibility, as well as a membrane-protecting effect, which underlines their good safety profile for potential biological intravenous application. Overall, CHEMSPOx, as a lipopolyoxazoline, holds great potential for versatile biological applications such as fast and direct intracellular delivery or cellular lysis protection.
Collapse
Affiliation(s)
| | - Liên Sabrina Reichel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | | | | | | | - Juliane Eberhardt
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter, Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter, Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes Christopher Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter, Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Marie Morille
- ICGM, CNRS, ENSCM, Univ. Montpellier, Montpellier, France
- Institut universitaire de France (IUF), Paris, France
| | | | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter, Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| |
Collapse
|
6
|
Greco G, Sarpietro MG. Liposome-Assisted Drug Delivery in the Treatment of Multiple Sclerosis. Molecules 2024; 29:4689. [PMID: 39407617 PMCID: PMC11477494 DOI: 10.3390/molecules29194689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the nervous system that leads to neurological dysfunctions and severe disabilities. It is worth noting that conventional pharmacotherapy is poorly selective and causes toxicity problems and several systemic side effects. Thus, there is a need to develop new approaches to this medical challenge. The use of nanocarriers for drug delivery represents a good strategy to overcome several issues such as high therapeutic drug doses with side effects, such as diarrhea, nausea, and abdominal pain, and drug degradation processes; in addition, nanocarriers can provide controlled and targeted drug release. This review describes the application of liposomes for the delivery of pharmaceutical actives to target MS. Firstly, MS is explained. Then, liposomes are described along with their preparation, characterization, and stability. The literature about the use of liposomes for the treatment of MS is then analyzed.
Collapse
Affiliation(s)
- Giuliana Greco
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
- NANOMED-Research Center on Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95125 Catania, Italy
| | - Maria Grazia Sarpietro
- NANOMED-Research Center on Nanomedicine and Pharmaceutical Nanotechnology, University of Catania, 95125 Catania, Italy
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| |
Collapse
|
7
|
Wang S, Yi K, Guan X, Zhou Z, Cao Y, Zhang X. Construction of charge-reversible coordination-crosslinked spherical nucleic acids to deliver dual anti-cancer genes and ferroptosis payloads. Int J Biol Macromol 2024; 277:134515. [PMID: 39106627 DOI: 10.1016/j.ijbiomac.2024.134515] [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: 05/10/2024] [Revised: 06/26/2024] [Accepted: 08/03/2024] [Indexed: 08/09/2024]
Abstract
Spherical nucleic acids (SNAs) are nanostructures with the DNA arranged radially on the surface, thus allowing specific binding with cancer cells expressing high levels of scavenger receptor-A to enhance cellular uptake. However, conventional carriers for SNAs are cytotoxic, not degradable and difficult to deliver multiple payloads. In this study, we developed charge-reversible coordination-crosslinked SNAs to deliver dual anti-cancer genes and ferroptosis payload for anti-cancer purposes. To this end, we modified poly(lactic acid) (PLA) with functionalized side chains to allow its binding with antisense oligonucleotides (ASOs) and siRNA, annealed two single-stranded RNAs to obtain double-stranded RNA, and introduced a polyethylene glycol (PEG) shell to enhance the circulation time. Additionally, the ferroptosis payload imidazole was coordinated with iron ions as a core-crosslinked group to enhance the stability of SNAs and efficiency to kill cancer cells. We demonstrated that this novel nanocomplex efficiently internalized and killed CT-26 cells in vitro. In vivo data confirmed that the dual gene delivery system successfully targeted CT-26 tumors in tumor-bearing BALB/c mice, and exhibited strong tumor suppression ability, without inducing adverse toxic effects. Taken together, our dual gene therapy system offered an enhanced anti-tumor solution by simultaneously delivering dual anti-cancer genes and ferroptosis payload in tumor microenvironment.
Collapse
Affiliation(s)
- Shuo Wang
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education and Key Laboratory of Polymeric Materials & Application Technology of Hunan Province, Xiangtan University, Xiangtan 411105, China
| | - Kailong Yi
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education and Key Laboratory of Polymeric Materials & Application Technology of Hunan Province, Xiangtan University, Xiangtan 411105, China
| | - Xiaoqi Guan
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education and Key Laboratory of Polymeric Materials & Application Technology of Hunan Province, Xiangtan University, Xiangtan 411105, China
| | - Zeyu Zhou
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education and Key Laboratory of Polymeric Materials & Application Technology of Hunan Province, Xiangtan University, Xiangtan 411105, China
| | - Yi Cao
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xuefei Zhang
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education and Key Laboratory of Polymeric Materials & Application Technology of Hunan Province, Xiangtan University, Xiangtan 411105, China.
| |
Collapse
|
8
|
Coorey CP, de Malmanche T, Chou A, Feddersen M. Complement-mediated thrombotic microangiopathy on a background of Alport syndrome: A case report. Nephrology (Carlton) 2024; 29:688-690. [PMID: 38604610 DOI: 10.1111/nep.14305] [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: 12/05/2023] [Revised: 02/10/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024]
Abstract
Here we present a case of complement-mediated thrombotic microangiopathy (TMA) in a patient who has a background of Stage 5 chronic kidney disease secondary to Alport syndrome. We explain our approach to the diagnosis of TMA, especially the reliance on non-renal manifestations of TMA and the role of kidney biopsy given there was a background of advanced kidney impairment at baseline.
Collapse
Affiliation(s)
- Craig Peter Coorey
- Renal Unit, Gosford Hospital, Gosford, New South Wales, Australia
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Theo de Malmanche
- Immunology, NSW Health Pathology, Newcastle, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Angela Chou
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Martin Feddersen
- Renal Unit, Gosford Hospital, Gosford, New South Wales, Australia
| |
Collapse
|
9
|
Mikled P, Chavasiri W, Khongkow M. Dual folate/biotin-decorated liposomes mediated delivery of methylnaphthazarin for anti-cancer activity. Sci Rep 2024; 14:21796. [PMID: 39294264 PMCID: PMC11410993 DOI: 10.1038/s41598-024-72532-7] [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: 06/20/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024] Open
Abstract
Chemotherapy is an effective strategy for mitigating the global challenge of cancer treatment, which often encounters drug resistance and negative side effects. Methylnaphthazarin (MNZ), a natural compound with promising anti-cancer properties, has been underexplored due to its poor aqueous solubility and low selectivity. This study introduces a novel approach to overcome these limitations by developing MNZ-encapsulating liposomes decorated with folate and biotin (F/B-LP-MNZ). This dual-targeting strategy aims to enhance the anti-cancer efficacy and specificity of MNZ delivery. Our innovative F/B-LP-MNZ formulation demonstrated excellent physicochemical properties, stability, and controlled drug release profiles. In vitro studies revealed that MNZ-loaded liposomes attenuate the toxicity associated with free MNZ while F/B-LP-MNZ significantly increased cytotoxicity against HeLa cells, which express high levels of folate and biotin receptors, compared to non-targeted liposomes. Enhanced cellular uptake and improved dynamic flow attachment further confirmed the superior specificity of F/B-LP in targeting cancer cells. Additionally, our results revealed that F/B-LP-MNZ effectively inhibits HeLa cell migration and adhesion through EMT suppression and apoptotic induction, indicating its potential to prevent cancer metastasis. These findings highlight the potential of dual folate and biotin receptors-targeting liposomes as an effective delivery system for MNZ, offering a promising new avenue for targeted cancer therapy.
Collapse
Affiliation(s)
- Pirun Mikled
- Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Warinthorn Chavasiri
- Center of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand.
| |
Collapse
|
10
|
Chen Z, Shu J, Hu Y, Mei H. Synergistic integration of mRNA-LNP with CAR-engineered immune cells: Pioneering progress in immunotherapy. Mol Ther 2024:S1525-0016(24)00605-1. [PMID: 39295145 DOI: 10.1016/j.ymthe.2024.09.019] [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: 04/26/2024] [Revised: 08/18/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has emerged as a revolutionary approach in the treatment of malignancies. Despite its remarkable successes, this field continues to grapple with challenges such as scalability, safety concerns, limited therapeutic effect, in vivo persistence, and the need for precise control over CAR expression. In the post-pandemic era of COVID-19 vaccine immunization, the application of messenger RNA (mRNA) encapsulated within lipid nanoparticles (LNPs) has recently garnered significant attention as a potential solution to address these challenges. This review delves into the dynamic landscape of mRNA-LNP technology and its potential implications for CAR-engineered immune cell-based immunotherapy.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Jinhui Shu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| |
Collapse
|
11
|
Rui X, Okamoto Y, Watanabe NM, Shimizu T, Wakileh W, Kajimura N, Umakoshi H. Preparation and characterization of macrophage membrane camouflaged cubosomes as a stabilized and immune evasive biomimetic nano-DDS. J Mater Chem B 2024; 12:8702-8715. [PMID: 39129447 DOI: 10.1039/d4tb01063a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
This study aims to develop a biomimetic nano-drug delivery system (nano-DDS) by employing a macrophage cell membrane camouflaging strategy to modify lyotropic liquid crystal nanoparticles (LLC-NPs). The cubic-structured LLC-NPs (Cubosomes, CBs) were prepared via a top-down approach (ultra-sonification) using monoolein (MO) and doped with the cationic lipid, DOTAP. The cell membrane camouflaging procedure induced changes in the cubic lipid phase from primitive cubic phase (QIIP) to a coexistence of QIIP and diamond cubic phase (QIID). The macrophage membrane camouflaging strategy protected CB cores from the destabilization by blood plasma and enhanced the stability of CBs. The in vitro experiment results revealed that the macrophage cell membrane coating significantly reduced macrophage uptake efficacy within 8 h of incubation compared to the non-camouflaged CBs, while it had minimal impact on cancer cell uptake efficacy. The macrophage membrane coated CBs showed lower accumulation in the heart, kidney and lungs in vivo. This study demonstrated the feasibility of employing cell membrane camouflaging on CBs and confirmed that the bio-functionalities of the CBs-based biomimetic nano-DDS were retained from the membrane source cells, and opened up promising possibilities for developing an efficient and safe drug delivery system based on the biomimetic approach.
Collapse
Affiliation(s)
- Xuehui Rui
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan.
| | - Yukihiro Okamoto
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan.
| | - Nozomi Morishita Watanabe
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan.
| | - Taro Shimizu
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ward Wakileh
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan.
| | - Naoko Kajimura
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, 7-1, Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Hiroshi Umakoshi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan.
| |
Collapse
|
12
|
Sharma S, Chakraborty M, Yadav D, Dhullap A, Singh R, Verma RK, Bhattacharya S, Singh S. Strategic Developments in Polymer-Functionalized Liposomes for Targeted Colon Cancer Therapy: An Updated Review of Clinical Trial Data and Future Horizons. Biomacromolecules 2024; 25:5650-5669. [PMID: 39162323 DOI: 10.1021/acs.biomac.4c00847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Liposomes, made up of phospholipid bilayers, are efficient nanocarriers for drug delivery because they can encapsulate both hydrophilic and lipophilic drugs. Conventional cancer treatments sometimes involve considerable toxicities and adverse drug reactions (ADRs), which limits their clinical value. Despite liposomes' promise in addressing these concerns, clinical trials have revealed significant limitations, including stability, targeted distribution, and scaling challenges. Recent clinical trials have focused on enhancing liposome formulations to increase therapeutic efficacy while minimizing negative effects. Notably, the approval of liposomal medications like Doxil demonstrates their potential in cancer treatment. However, the intricacy of liposome preparation and the requirement for comprehensive regulatory approval remain substantial impediments. Current clinical trial updates show continued efforts to improve liposome stability, targeting mechanisms, and payload capacity in order to address these issues. The future of liposomal drug delivery in cancer therapy depends on addressing these challenges in order to provide patients with more effective and safer treatment alternatives.
Collapse
Affiliation(s)
- Satyam Sharma
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| | - Moitrai Chakraborty
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| | - Dharmendra Yadav
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| | - Aniket Dhullap
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| | - Raghuraj Singh
- Pharmaceutical Nanotechnology lab, Institute of Nano Science and Technology (INST), Sector 81, Mohali, Punjab 140306, India
| | - Rahul Kumar Verma
- Pharmaceutical Nanotechnology lab, Institute of Nano Science and Technology (INST), Sector 81, Mohali, Punjab 140306, India
| | - Sankha Bhattacharya
- SVKM's NMIMS School of Pharmacy & Technology Management, Shirpur, Dist. Dhule, Maharashtra 425405, India
| | - Sanjiv Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur 844102, India
| |
Collapse
|
13
|
Zhu X, Luo W, Zhang D, Liu R. An Assay for Immunogenic Detection of Anti-PEG Antibody. Chembiochem 2024; 25:e202400316. [PMID: 38867605 DOI: 10.1002/cbic.202400316] [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: 04/05/2024] [Revised: 05/24/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024]
Abstract
With the increasing use of polyethylene glycol (PEG) based proteins and drug delivery systems, anti-PEG antibodies have commonly been detected among the population, causing the accelerated blood clearance and hypersensitivity reactions, poses potential risks to the clinical efficacy and safety of PEGylated drugs. Therefore, vigilant monitoring of anti-PEG antibodies is crucial for both research and clinical guidance regarding PEGylated drugs. The enzyme-linked immunosorbent assay (ELISA) is a common method for detecting anti-PEG antibodies. However, diverse coating methods, blocking solutions and washing solutions have been employed across different studies, and unsuitable use of Tween 20 as the surfactant even caused biased results. In this study, we established the optimal substrate coating conditions, and investigated the influence of various surfactants and blocking solutions on the detection accuracy. The findings revealed that incorporating 1 % bovine serum albumin into the serum dilution in the absence of surfactants will result the credible outcomes of anti-PEG antibody detection.
Collapse
Affiliation(s)
- Xiang Zhu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Weizhe Luo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Donghui Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
- School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
14
|
Laha A, Nasra S, Bhatia D, Kumar A. Advancements in rheumatoid arthritis therapy: a journey from conventional therapy to precision medicine via nanoparticles targeting immune cells. NANOSCALE 2024; 16:14975-14993. [PMID: 39056352 DOI: 10.1039/d4nr02182g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Rheumatoid arthritis (RA) is a progressive autoimmune disease that mainly affects the inner lining of the synovial joints and leads to chronic inflammation. While RA is not known as lethal, recent research indicates that it may be a silent killer because of its strong association with an increased risk of chronic lung and heart diseases. Patients develop these systemic consequences due to the regular uptake of heavy drugs such as disease-modifying antirheumatic medications (DMARDs), glucocorticoids (GCs), nonsteroidal anti-inflammatory medicines (NSAIDs), etc. Nevertheless, a number of these medications have off-target effects, which might cause adverse toxicity, and have started to become resistant in patients as well. Therefore, alternative and promising therapeutic techniques must be explored and adopted, such as post-translational modification inhibitors (like protein arginine deiminase inhibitors), RNA interference by siRNA, epigenetic drugs, peptide therapy, etc., specifically in macrophages, neutrophils, Treg cells and dendritic cells (DCs). As the target cells are specific, ensuring targeted delivery is also equally important, which can be achieved with the advent of nanotechnology. Furthermore, these nanocarriers have fewer off-site side effects, enable drug combinations, and allow for lower drug dosages. Among the nanoparticles that can be used for targeting, there are both inorganic and organic nanomaterials such as solid-lipid nanoparticles, liposomes, hydrogels, dendrimers, and biomimetics that have been discussed. This review highlights contemporary therapy options targeting macrophages, neutrophils, Treg cells, and DCs and explores the application of diverse nanotechnological techniques to enhance precision RA therapies.
Collapse
Affiliation(s)
- Anwesha Laha
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Simran Nasra
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Dhiraj Bhatia
- Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar - 382055, Gujarat, India
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| |
Collapse
|
15
|
Maiti D, Yokoyama M, Shiraishi K. Impact of the Hydrophilicity of Poly(sarcosine) on Poly(ethylene glycol) (PEG) for the Suppression of Anti-PEG Antibody Binding. ACS OMEGA 2024; 9:34577-34588. [PMID: 39157078 PMCID: PMC11325419 DOI: 10.1021/acsomega.4c02655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 08/20/2024]
Abstract
A method of poly(ethylene glycol) (PEG) conjugation is known as PEGylation, which has been employed to deliver therapeutic drugs, proteins, or nanoparticles by considering the intrinsic non- or very low immunogenic property of PEG. However, PEG has its weaknesses, and one major concern is the potential immunogenicity of PEGylated proteins. Because of its hydrophilicity, poly(sarcosine) (P(Sar)) may be an attractive-and superior-substitute for PEG. In the present study, we designed a double hydrophilic diblock copolymer, methoxy-PEG-b-P(Sar) m (m = 5-55) (mPEG-P(Sar) m ), and synthesized a triblock copolymer with hydrophobic poly(l-isoleucine) (P(Ile)). We validated that double hydrophilic mPEG-P(Sar) block copolymers suppressed the specific binding of three monoclonal anti-PEG antibodies (anti-PEG mAbs) to PEG. The results of our indirect ELISAs indicate that P(Sar) significantly helps to reduce the binding of anti-PEG mAbs to PEG. Importantly, the steady suppression of this binding was made possible, in part, thanks to the maximum number of sarcosine units in the triblock copolymer, as evidenced by sandwich ELISA and biolayer interferometry assay (BLI): the intrinsic hydrophilicity of P(Sar) had a clear supportive effect on PEG. Finally, because we used P(Ile) as a hydrophobic block, PEG-P(Sar) might be an attractive alternative to PEG in the search for protein shields that minimize the immunogenicity of PEGylated proteins.
Collapse
Affiliation(s)
- Debabrata Maiti
- Research Center for Medical
Sciences, The Jikei University School of
Medicine, 163-1, Kashiwa-shita, Kashiwa, Chiba 277-0004, Japan
| | - Masayuki Yokoyama
- Research Center for Medical
Sciences, The Jikei University School of
Medicine, 163-1, Kashiwa-shita, Kashiwa, Chiba 277-0004, Japan
| | - Kouichi Shiraishi
- Research Center for Medical
Sciences, The Jikei University School of
Medicine, 163-1, Kashiwa-shita, Kashiwa, Chiba 277-0004, Japan
| |
Collapse
|
16
|
Bai X, Chen Q, Li F, Teng Y, Tang M, Huang J, Xu X, Zhang XQ. Optimized inhaled LNP formulation for enhanced treatment of idiopathic pulmonary fibrosis via mRNA-mediated antibody therapy. Nat Commun 2024; 15:6844. [PMID: 39122711 PMCID: PMC11315999 DOI: 10.1038/s41467-024-51056-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Lipid nanoparticle-assisted mRNA inhalation therapy necessitates addressing challenges such as resistance to shear force damage, mucus penetration, cellular internalization, rapid lysosomal escape, and target protein expression. Here, we introduce the innovative "LOOP" platform with a four-step workflow to develop inhaled lipid nanoparticles specifically for pulmonary mRNA delivery. iLNP-HP08LOOP featuring a high helper lipid ratio, acidic dialysis buffer, and excipient-assisted nebulization buffer, demonstrates exceptional stability and enhanced mRNA expression in the lungs. By incorporating mRNA encoding IL-11 single chain fragment variable (scFv), scFv@iLNP-HP08LOOP effectively delivers and secretes IL-11 scFv to the lungs of male mice, significantly inhibiting fibrosis. This formulation surpasses both inhaled and intravenously injected IL-11 scFv in inhibiting fibroblast activation and extracellular matrix deposition. The HP08LOOP system is also compatible with commercially available ALC0315 LNPs. Thus, the "LOOP" method presents a powerful platform for developing inhaled mRNA nanotherapeutics with potential for treating various respiratory diseases, including idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Xin Bai
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Qijing Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Fengqiao Li
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Yilong Teng
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Maoping Tang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Huang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyang Xu
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA.
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA.
| | - Xue-Qing Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China.
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
17
|
Ladva DN, Selvadoss PP, Chitroda GK, Dhanasekaran S, Nellore J, Tippabathani J, Solomon SM. Maleimide conjugated PEGylated liposomal antibiotic to combat multi-drug resistant Escherichia coli and Klebsiella pneumoniae with enhanced wound healing potential. Sci Rep 2024; 14:18361. [PMID: 39112534 PMCID: PMC11306640 DOI: 10.1038/s41598-024-68647-6] [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: 05/16/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Antibiotic resistance is a significant threat, leaving us vulnerable to bacterial infections. Novel strategies are needed to combat bacterial resistance beyond discovering new antibiotics. This research focuses on using maleimide conjugated PEGylated liposomes (Mal-PL-Ab) to individually encapsulate a variety of antibiotics (ceftriaxone, cephalexin, doxycycline, piperacillin, ampicillin, and ceftazidime) and enhance their delivery against multi-drug resistant (MDR) bacteria like Escherichia coli (E. coli) and Klebsiella pneumoniae (K. pneumoniae). Mal-PL-Ab, with an average size of 84.2 nm ± 4.32 nm, successfully encapsulated these antibiotics with an encapsulation efficiency of 37.73 ± 3.19%. Compared to non-PEGylated liposomes (L-Ab), Mal-PL-Ab exhibited reduced toxicity in human dermal cells, emphasizing the importance of PEGylation in minimizing adverse effects. Mal-PL-Ab significantly decreased the minimum inhibitory concentration (MIC) values against both E. coli and K. pneumoniae by 9.33-fold and eightfold reduction (compared to non-PEGylated liposomes with 2.33-fold and 2.33fold reduction), respectively, indicating enhanced efficacy against MDR strains. Furthermore, in vitro scratch assay and gene expression analysis of human dermal fibroblast revealed that Mal-PL-Ab promoted cell proliferation, migration, and wound healing through upregulation of cell cycle, DNA repair, and angiogenesis-related genes. Harnessing the power of encapsulation, Mal-PL-Ab presents a novel avenue for enhanced antibiotic delivery and wound healing, potentially transcending the limitations of traditional options.
Collapse
Affiliation(s)
- Darshan Narendrabhai Ladva
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, Gujarat, 382426, India
| | - Pradeep Pushparaj Selvadoss
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, Gujarat, 382426, India.
| | - Grishma Kantibhai Chitroda
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, Gujarat, 382426, India
| | - Sivaraman Dhanasekaran
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, Gujarat, 382426, India
| | - Jayshree Nellore
- Department of Biotechnology, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | | | - Sundar Manoharan Solomon
- Department of Chemistry, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, Gujarat, 382426, India.
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, Gujarat, 382426, India.
| |
Collapse
|
18
|
Cao X, Li K, Wang J, Xie X, Sun L. PBPK model of pegylated liposomal doxorubicin to simultaneously predict the concentration-time profile of encapsulated and free doxorubicin in tissues. Drug Deliv Transl Res 2024:10.1007/s13346-024-01680-0. [PMID: 39103592 DOI: 10.1007/s13346-024-01680-0] [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] [Accepted: 07/20/2024] [Indexed: 08/07/2024]
Abstract
The objective of this study was to develop a physiologically based pharmacokinetic (PBPK) model to predict the concentrations of encapsulated and free doxorubicin in plasma and tissues in mice after intravenous injection of PEGylated liposomes (Doxil®). The PBPK model used in this study contains liposomes and free doxorubicin disposition components. The free doxorubicin disposition component was used to simulate the disposition of free doxorubicin produced by mononuclear phagocyte system (MPS)-degrading liposomes. The liver, spleen, kidneys, and lungs contain an additional MPS subcompartment. These compartments are interconnected through blood and lymphatic circulation. The model was validated strictly by four doses of external observed plasma and tissue concentration-time profiles. The fold error (FE) values were almost all within threefold. The sensitivity analysis revealed that the MPS-related parameters greatly influenced the model. The predicted in vivo distribution characteristics of the doxorubicin liposomes and doxorubicin solution were consistent with the observed values. The PBPK model was established based on the physiological mechanism and parameters of practical significance that can be measured in vitro. Thus, it can be used to study the pharmacokinetic properties of liposomes. This study also provides a reference for the establishment of liposome PBPK model.
Collapse
Affiliation(s)
- Xuewei Cao
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Kejun Li
- China Medical University-The Queen's University of Belfast Joint College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Jingyu Wang
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Xiaoqian Xie
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Le Sun
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| |
Collapse
|
19
|
Li Y, Xin X, Zhou X, Liu J, Liu H, Yuan S, Liu H, Hao W, Sun J, Wang Y, Gong W, Yang M, Li Z, Han Y, Gao C, Yang Y. ROS-responsive biomimetic nanosystem camouflaged by hybrid membranes of platelet-exosomes engineered with neuronal targeting peptide for TBI therapy. J Control Release 2024; 372:531-550. [PMID: 38851535 DOI: 10.1016/j.jconrel.2024.06.018] [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: 01/03/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Recovery and survival following traumatic brain injury (TBI) depends on optimal amelioration of secondary injuries at lesion site. Delivering mitochondria-protecting drugs to neurons may revive damaged neurons at sites secondarily traumatized by TBI. Pioglitazone (PGZ) is a promising candidate for TBI treatment, limited by its low brain accumulation and poor targetability to neurons. Herein, we report a ROS-responsive nanosystem, camouflaged by hybrid membranes of platelets and engineered extracellular vesicles (EVs) (C3-EPm-|TKNPs|), that can be used for targeted delivery of PGZ for TBI therapy. Inspired by intrinsic ability of macrophages for inflammatory chemotaxis, engineered M2-like macrophage-derived EVs were constructed by fusing C3 peptide to EVs membrane integrator protein, Lamp2b, to confer them with ability to target neurons in inflamed lesions. Platelets provided hybridized EPm with capabilities to target hemorrhagic area caused by trauma via surface proteins. Consequently, C3-EPm-|PGZ-TKNPs| were orientedly delivered to neurons located in the traumatized hemisphere after intravenous administration, and triggered the release of PGZ from TKNPs via oxidative stress. The current work demonstrate that C3-EPm-|TKNPs| can effectively deliver PGZ to alleviate mitochondrial damage via mitoNEET for neuroprotection, further reversing behavioral deficits in TBI mice. Our findings provide proof-of-concept evidence of C3-EPm-|TKNPs|-derived nanodrugs as potential clinical approaches against neuroinflammation-related intracranial diseases.
Collapse
Affiliation(s)
- Yi Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Xin Xin
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Xun Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; College of Pharmacy, Henan University, Kaifeng 475000, People's Republic of China
| | - Jingzhou Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Hangbing Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Shuo Yuan
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China; School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Hanhan Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Wenyan Hao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Jiejie Sun
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Yuli Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Wei Gong
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Meiyan Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Zhiping Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China
| | - Yang Han
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Chunsheng Gao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China.
| | - Yang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, People's Republic of China.
| |
Collapse
|
20
|
Eker F, Duman H, Akdaşçi E, Bolat E, Sarıtaş S, Karav S, Witkowska AM. A Comprehensive Review of Nanoparticles: From Classification to Application and Toxicity. Molecules 2024; 29:3482. [PMID: 39124888 PMCID: PMC11314082 DOI: 10.3390/molecules29153482] [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: 07/03/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Nanoparticles are structures that possess unique properties with high surface area-to-volume ratio. Their small size, up to 100 nm, and potential for surface modifications have enabled their use in a wide range of applications. Various factors influence the properties and applications of NPs, including the synthesis method and physical attributes such as size and shape. Additionally, the materials used in the synthesis of NPs are primary determinants of their application. Based on the chosen material, NPs are generally classified into three categories: organic, inorganic, and carbon-based. These categories include a variety of materials, such as proteins, polymers, metal ions, lipids and derivatives, magnetic minerals, and so on. Each material possesses unique attributes that influence the activity and application of the NPs. Consequently, certain NPs are typically used in particular areas because they possess higher efficiency along with tenable toxicity. Therefore, the classification and the base material in the NP synthesis hold significant importance in both NP research and application. In this paper, we discuss these classifications, exemplify most of the major materials, and categorize them according to their preferred area of application. This review provides an overall review of the materials, including their application, and toxicity.
Collapse
Affiliation(s)
- Furkan Eker
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Emir Akdaşçi
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Ecem Bolat
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Sümeyye Sarıtaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (F.E.); (H.D.); (E.A.); (E.B.); (S.S.)
| | - Anna Maria Witkowska
- Department of Food Biotechnology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
21
|
Karahmet Sher E, Alebić M, Marković Boras M, Boškailo E, Karahmet Farhat E, Karahmet A, Pavlović B, Sher F, Lekić L. Nanotechnology in medicine revolutionizing drug delivery for cancer and viral infection treatments. Int J Pharm 2024; 660:124345. [PMID: 38885775 DOI: 10.1016/j.ijpharm.2024.124345] [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: 02/26/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Advancements in nanotechnology were vastly applied in medicine and pharmacy, especially in the field of nano-delivery systems. It took a long time for these systems to ensure precise delivery of very delicate molecules, such as RNA, to cells at concentrations that yield remarkable efficiency, with success rates reaching 95.0% and 94.5%. These days, there are several advantages of using nanotechnological solutions in the prevention and treatment of cancer and viral infections. Its interventions improve treatment outcomes both due to increased effectiveness of the drug at target location and by reducing adverse reactions, thereby increasing patient adherence to the therapy. Based on the current knowledge an updated review was made, and perspective, opportunities and challenges in nanomedicine were discussed. The methods employed include comprehensive examination of existing literature and studies on nanoparticles and nano-delivery systems including both in vitro tests performed on cell cultures and in vivo assessments carried out on appropriate animal models, with a specific emphasis on their applications in oncology and virology. This brings together various aspects including both structure and formation as well as its association with characteristic behaviour in organisms, providing a novel perspective. Furthermore, the practical application of these systems in medicine and pharmacy with a focus on viral diseases and malignancies was explored. This review can serve as a valuable guide for fellow researchers, helping them navigate the abundance of findings in this field. The results indicate that applications of nanotechnological solutions for the delivery of medicinal products improving therapeutic outcomes will continue to expand.
Collapse
Affiliation(s)
- Emina Karahmet Sher
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom.
| | - Mirna Alebić
- Department of Pharmacy, University Hospital Centre Zagreb, Zagreb 10000, Croatia
| | - Marijana Marković Boras
- Department of Laboratory Diagnostic, University Clinical Hospital Mostar, Mostar 88000, Bosnia and Herzegovina; International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Emina Boškailo
- International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Esma Karahmet Farhat
- International Society of Engineering Science and Technology, Nottingham, United Kingdom; Department of Food and Nutrition, Faculty of Food Technology, Juraj Strossmayer University of Osijek, Osijek 31000, Croatia
| | - Alma Karahmet
- International Society of Engineering Science and Technology, Nottingham, United Kingdom
| | - Bojan Pavlović
- Faculty of Physical Education and Sports, University of East Sarajevo, Lukavica, Republika Srpska 75327, Bosnia and Herzegovina
| | - Farooq Sher
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom.
| | - Lana Lekić
- Faculty of Health Studies, University of Sarajevo, Sarajevo 71000, Bosnia and Herzegovina
| |
Collapse
|
22
|
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
|
23
|
Masclef JB, Acs EMN, Koehnke J, Prunet J, Schmidt BVKJ. PEGose Block Poly(lactic acid) Nanoparticles for Cargo Delivery. Macromolecules 2024; 57:6013-6023. [PMID: 39005948 PMCID: PMC11238580 DOI: 10.1021/acs.macromol.4c00528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/15/2024] [Accepted: 06/04/2024] [Indexed: 07/16/2024]
Abstract
Hydrophilic polymers have found ubiquitous use in drug delivery and novel polymer materials to advance drug delivery systems are highly sought after. Herein, an amylose mimic (PEGose) was combined with poly(lactic acid) (PLA) in an amphiphilic block copolymer to form PEG-free nanoparticles as an alternative to PEG-based nanomedicines. The block copolymer self-assembled into 150-200 nm particles with a narrow dispersity in aqueous environment. The formed nanoparticles were capable of encapsulation, the sustained release of both hydrophilic and hydrophobic dyes. Moreover, the nanoparticles were found to be remarkably stable and had a very low cytotoxicity and a high propensity to penetrate cells. These results highlight the potential of PEGose-b-PLA to be used in drug delivery with a new hydrophilic building block.
Collapse
Affiliation(s)
- Jean-Baptiste Masclef
- School
of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ Glasgow, U.K.
| | - Emmanuelle M. N. Acs
- School
of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ Glasgow, U.K.
| | - Jesko Koehnke
- School
of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ Glasgow, U.K.
- Institute
of Food Chemistry, Leibniz University Hannover, 30167 Hannover, Germany
| | - Joëlle Prunet
- School
of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ Glasgow, U.K.
| | | |
Collapse
|
24
|
Wei L, Lin L, Wang J, Guan X, Li W, Gui Y, Liao S, Wang M, Li J, Deng Y, Song Y. The selection of animal models influences the assessment of anti-tumor efficacy: promising sialic acid-conjugate modified liposomes demonstrate remarkable therapeutic effects in diverse mouse strains. Drug Deliv Transl Res 2024; 14:1794-1809. [PMID: 38165530 DOI: 10.1007/s13346-023-01502-9] [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] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
Mice as a crucial tool for preclinical assessment of antineoplastic agents. The impact of physiological differences among mouse strains on the in vivo efficacy of antitumor drugs, however, has been significantly overlooked. Mononuclear phagocyte system (MPS) is the major player in clearance in vivo, and differences in MPS among different strains may potentially impact the effectiveness of antitumor preparations. Therefore, in this study, we employed conventional liposomes (CL-EPI) and SA-ODA modified liposomes (SAL-EPI) as model preparations to investigate the comprehensive tumor therapeutic effects of CL-EPI and SAL-EPI in KM, BALB/c, and C57BL/6 tumor-bearing mice. The results demonstrated significant variability in the efficacy of CL-EPI for tumor treatment across different mouse strains. Therefore, we should pay attention to the selection of animal models in the study of antitumor agents. SAL-EPI effectively targeted tumor sites by binding to Siglec-1 on the surface of peripheral blood monocytes (PBMs), and achieved good therapeutic effect in different mouse strains with little difference in treatment. The SA modified preparation is therefore expected to achieve a favorable therapeutic effect in tumor patients with different immune states through PBMs delivery (Siglec-1 was expressed in both mice and humans), thereby possessing clinical translational value and promising development prospects.
Collapse
Affiliation(s)
- Lu Wei
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Lin Lin
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Jia Wang
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Xinying Guan
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Wen Li
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Yangxu Gui
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Shupei Liao
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Mingyang Wang
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Jiaqi Li
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, China.
| |
Collapse
|
25
|
Wang Y, Liu C, Ren Y, Song J, Fan K, Gao L, Ji X, Chen X, Zhao H. Nanomaterial-Based Strategies for Attenuating T-Cell-Mediated Immunodepression in Stroke Patients: Advancing Research Perspectives. Int J Nanomedicine 2024; 19:5793-5812. [PMID: 38882535 PMCID: PMC11180442 DOI: 10.2147/ijn.s456632] [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: 12/25/2023] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
This review article discusses the potential of nanomaterials in targeted therapy and immunomodulation for stroke-induced immunosuppression. Although nanomaterials have been extensively studied in various biomedical applications, their specific use in studying and addressing immunosuppression after stroke remains limited. Stroke-induced neuroinflammation is characterized by T-cell-mediated immunodepression, which leads to increased morbidity and mortality. Key observations related to immunodepression after stroke, including lymphopenia, T-cell dysfunction, regulatory T-cell imbalance, and cytokine dysregulation, are discussed. Nanomaterials, such as liposomes, micelles, polymeric nanoparticles, and dendrimers, offer advantages in the precise delivery of drugs to T cells, enabling enhanced targeting and controlled release of immunomodulatory agents. These nanomaterials have the potential to modulate T-cell function, promote neuroregeneration, and restore immune responses, providing new avenues for stroke treatment. However, challenges related to biocompatibility, stability, scalability, and clinical translation need to be addressed. Future research efforts should focus on comprehensive studies to validate the efficacy and safety of nanomaterial-based interventions targeting T cells in stroke-induced immunosuppression. Collaborative interdisciplinary approaches are necessary to advance the field and translate these innovative strategies into clinical practice, ultimately improving stroke outcomes and patient care.
Collapse
Grants
- This work was supported by the National Natural Science Foundation of China (Grant number 82001248), National University of Singapore (NUHSRO/2020/133/Startup/08, NUHSRO/2023/008/NUSMed/TCE/LOA, NUHSRO/2021/034/TRP/09/Nanomedicine, NUHSRO/2021/044/Kickstart/09/LOA, 23-0173-A0001), National Medical Research Council (MOH-001388-00, CG21APR1005, OFIRG23jul-0047), Singapore Ministry of Education (MOE-000387-00), and National Research Foundation (NRF-000352-00)
Collapse
Affiliation(s)
- Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Yanhong Ren
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Heng Zhao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
26
|
Sameer Khan M, Gupta G, Alsayari A, Wahab S, Sahebkar A, Kesharwani P. Advancements in liposomal formulations: A comprehensive exploration of industrial production techniques. Int J Pharm 2024; 658:124212. [PMID: 38723730 DOI: 10.1016/j.ijpharm.2024.124212] [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: 03/12/2024] [Revised: 04/24/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
Liposomes are nanosized, spherical vesicles consisting of an aqueous core encircled by one or more phospholipid bilayer shells. Liposomes have found extensive use in numerous biomedicine and nanomedicine applications due to their excellent biocompatibility, adaptable chemical composition, ease of preparation, and diverse structural characteristics. These applications include nanocarriers for drug delivery, immunoassays, nutraceuticals, tissue engineering, clinical diagnostics, and theranostics formulations. These applications stimulated significant efforts toward scaling up formation processes in anticipation of appropriate industrial advancement. Despite the advancements in conventional methods and the emergence of new approaches for liposome production, their inherent susceptibility to chemical and mechanical influences contributes to critical challenges, including limited colloidal stability and decreased efficiency in encapsulating cargo molecules. With this context, the current review provides brief insights into liposomes conventional and novel industrial production techniques. With a special focus on the structural parameters, and pivotal elements influencing the synthesis of an appropriate and stable formulation, followed by the various regulatory aspects of industrial production.
Collapse
Affiliation(s)
- Mohammad Sameer Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Garima Gupta
- Graphic Era Hill University, Dehradun 248002, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
27
|
Soto-Arriaza M, Cena Ahumada E, Bonardd S, Melendez J. Calcein release from DPPC liposomes by phospholipase A2 activity: Effect of cholesterol and amphipathic copolymers. J Liposome Res 2024:1-13. [PMID: 38850012 DOI: 10.1080/08982104.2024.2361610] [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: 08/02/2023] [Accepted: 05/25/2024] [Indexed: 06/09/2024]
Abstract
In this study, we evaluated the impact of incorporating diblock and triblock amphiphilic copolymers, as well as cholesterol into DPPC liposomes on the release of a model molecule, calcein, mediated by exogenous phospholipase A2 activity. Our findings show that calcein release slows down in the presence of copolymers at low concentration, while at high concentration, the calcein release profile resembles that of the DPPC control. Additionally, calcein release mediated by exogenous PLA2 decreases as the amount of solubilized cholesterol increases, with a maximum between 18 mol% and 20 mol%. At concentrations higher than 24 mol%, no calcein release was observed. Studies conducted on HEK-293 and HeLa cells revealed that DPPC liposomes reduced viability by only 5% and 12%, respectively, after 3 hours of incubation, while DPPC liposome in presence of 33 mol% of Cholesterol reduced viability by approximately 11% and 23%, respectively, during the same incubation period. For formulations containing copolymers at low and high concentrations, cell viability decreased by approximately 20% and 40%, respectively, after 3 hours of incubation. Based on these preliminary results, we can conclude that the presence of amphiphilic copolymers at low concentration can be used in the design of new DPPC liposomes, and together with cholesterol, they can modulate liposome stabilization. The new formulations showed low cytotoxicity in HEK-293 cells, and it was observed that calcein release depended entirely on PLA2 activity and the presence of calcium ions.
Collapse
Affiliation(s)
- Marco Soto-Arriaza
- Escuela de Química y Farmacia, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Eduardo Cena Ahumada
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Sebastián Bonardd
- Centro de Física de Materiales (CSIC, UPV/EHU)-Materials Physics Center (MPC), Donostia-San Sebastían, Spain
- Department of Polymers and Advanced Materials: Physics, Chemistry and Technology, University of the Basque Country UPV/EHU, Donostia-San Sebastian, Spain
| | | |
Collapse
|
28
|
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
|
29
|
Sanaee M, Ronquist KG, Sandberg E, Morrell JM, Widengren J, Gallo K. Antibody-Loading of Biological Nanocarrier Vesicles Derived from Red-Blood-Cell Membranes. ACS OMEGA 2024; 9:22711-22718. [PMID: 38826552 PMCID: PMC11137724 DOI: 10.1021/acsomega.4c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 06/04/2024]
Abstract
Antibodies, disruptive potent therapeutic agents against pharmacological targets, face a barrier in crossing immune systems and cellular membranes. To overcome these, various strategies have been explored including shuttling via liposomes or biocamouflaged nanoparticles. Here, we demonstrate the feasibility of loading antibodies into exosome-mimetic nanovesicles derived from human red-blood-cell membranes, which can act as nanocarriers for intracellular delivery. Goat-antichicken antibodies are loaded into erythrocyte-derived nanovesicles, and their loading yields are characterized and compared with smaller dUTP-cargo molecules. Applying dual-color coincident fluorescence burst analyses, the loading yield of nanocarriers is rigorously profiled at the single-vesicle level, overcoming challenges due to size-heterogeneity and demonstrating a maximum antibody-loading yield of 38-41% at the optimal vesicle radius of 52 nm. The achieved average loading yields, amounting to 14% across the entire nanovesicle population, with more than two antibodies per loaded vesicle, are fully comparable to those obtained for the much smaller dUTP molecules loaded in the nanovesicles after additional exosome-spin-column purification. The results suggest a promising new avenue for therapeutic delivery of antibodies, potentially encompassing also intracellular targets and suitable for large-scale pharmacological applications, which relies on the exosome-mimetic properties, biocompatibility, and low-immunogenicity of bioengineered nanocarriers synthesized from human erythrocyte membranes.
Collapse
Affiliation(s)
- Maryam Sanaee
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm 10691, Sweden
| | - K. Göran Ronquist
- Department
of Clinical Sciences, Swedish University
of Agricultural Sciences, Uppsala 75007, Sweden
| | - Elin Sandberg
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm 10691, Sweden
| | - Jane M. Morrell
- Department
of Clinical Sciences, Swedish University
of Agricultural Sciences, Uppsala 75007, Sweden
| | - Jerker Widengren
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm 10691, Sweden
| | - Katia Gallo
- Department
of Applied Physics, School of Engineering Sciences, KTH Royal Institute of Technology, Stockholm 10691, Sweden
| |
Collapse
|
30
|
Lee Y, Jeong M, Lee G, Park J, Jung H, Im S, Lee H. Development of Lipid Nanoparticle Formulation for the Repeated Administration of mRNA Therapeutics. Biomater Res 2024; 28:0017. [PMID: 38779139 PMCID: PMC11109479 DOI: 10.34133/bmr.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/13/2024] [Indexed: 05/25/2024] Open
Abstract
During the COVID-19 pandemic, mRNA vaccines emerged as a rapid and effective solution for global immunization. The success of COVID-19 mRNA vaccines has increased interest in the use of lipid nanoparticles (LNPs) for the in vivo delivery of mRNA therapeutics. Although mRNA exhibits robust expression profiles, transient protein expression is often observed, raising uncertainty regarding the frequency of its administration. Additionally, various RNA therapeutics may necessitate repeated dosing to achieve optimal therapeutic outcomes. Nevertheless, the impact of repeated administrations of mRNA/LNP on immune responses and protein expression efficacy remains unclear. In this study, we investigated the influence of the formulation parameters, specifically ionizable lipids and polyethylene glycol (PEG) lipids, on the repeat administration of mRNA/LNP. Our findings revealed that ionizable lipids had no discernible impact on the dose-responsive efficacy of repeat administrations, whereas the lipid structure and molar ratio of PEG lipids were primary factors that affected mRNA/LNP performance. The optimization of the LNP formulation with PEG lipid confirmed the sustained dose-responsive efficacy of mRNA after repeated administrations. This study highlights the critical importance of optimizing LNP formulations for mRNA therapeutics requiring repeated administrations.
Collapse
Affiliation(s)
- Yeji Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Michaela Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Gyeongseok Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jeongeun Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hyein Jung
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seongeun Im
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences,
Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
31
|
Pinto M, Machado CS, Barreiro S, Otero-Espinar FJ, Remião F, Borges F, Fernandes C. Rescuing a Troubled Tolcapone with PEGylated PLGA Nanoparticles: Design, Characterization, and Hepatotoxicity Evaluation. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21522-21533. [PMID: 38647198 DOI: 10.1021/acsami.4c00614] [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: 04/25/2024]
Abstract
Tolcapone is an orally active catechol-O-methyltransferase (COMT) inhibitor used as adjuvant therapy in Parkinson's disease. However, it has a highly hepatotoxic profile, as recognized by the U.S. Food and Drug Administration. As a possible solution, nanoscience brought us several tools in the development of new functional nanomaterials with tunable physicochemical properties, which can be part of a solution to solve several drawbacks, including drug's short half-life and toxicity. This work aims to use PEGylated poly(lactic-co-glycolic acid) (PLGA) nanoparticles as a stable carrier with lower hydrodynamic size and polydispersity to encapsulate tolcapone in order to overcome its therapeutic drawbacks. Using the nanoprecipitation method, tolcapone-loaded nanoparticles with a DLC% of 5.7% were obtained (EE% of 47.0%) and subjected to a lyophilization optimization process to obtain a final shelf-stable formulation. Six different cryoprotectants in concentrations up to 10% (w/v) were tested. A formulation of PLGA nanoparticles with 3% hydroxypropyl-β-cyclodextrin (HPβCD) as a cryoprotectant (PLGA-HP@Tolc), presenting sub-200 nm sizes and low polydispersity (PdI < 0.200) was selected. Cytotoxicity assays, namely, MTT and SRB, were used to study the metabolic activity and cell density of tolcapone and PLGA-HP@Tolc-treated cells. In both assays, a hepatocarcinoma cell line (HepG2) growing in glucose or glucose-free media (galactose-supplemented medium) was used. The results demonstrated that the treatment with the PLGA-HP@Tolc formulation led to a decrease in cytotoxicity in comparison to free tolcapone-treated cells in both media tested. Moreover, the elected formulation also counteracted ATP-depletion and excessive ROS production induced by tolcapone. The results suggest that HPβCD might have a dual function in the formulation: cryoprotectant and anticytotoxic agent, protecting cells from tolcapone-induced damage. Using an in vitro COMT inhibition assay, the PLGA-HP@Tolc formulation demonstrated to inhibit COMT as efficiently as free tolcapone. Overall, the results suggest that tolcapone-loaded PLGA NPs could be an interesting alternative to free tolcapone, demonstrating the same in vitro efficacy in inhibiting COMT but with a safer cytotoxic profile.
Collapse
Affiliation(s)
- Miguel Pinto
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, Porto 4169-007, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
| | - Cláudia Sofia Machado
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, Porto 4169-007, Portugal
| | - Sandra Barreiro
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, Porto 4169-007, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
| | - Francisco J Otero-Espinar
- Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy, University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Fernando Remião
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, R. Jorge de Viterbo Ferreira 228, Porto 4050-313, Portugal
| | - Fernanda Borges
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, Porto 4169-007, Portugal
| | - Carlos Fernandes
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R.Campo Alegre s/n, Porto 4169-007, Portugal
| |
Collapse
|
32
|
Zou Y, Kamoi K, Zong Y, Zhang J, Yang M, Ohno-Matsui K. Vaccines and the Eye: Current Understanding of the Molecular and Immunological Effects of Vaccination on the Eye. Int J Mol Sci 2024; 25:4755. [PMID: 38731972 PMCID: PMC11084287 DOI: 10.3390/ijms25094755] [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: 03/23/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Vaccination is a public health cornerstone that protects against numerous infectious diseases. Despite its benefits, immunization implications on ocular health warrant thorough investigation, particularly in the context of vaccine-induced ocular inflammation. This review aimed to elucidate the complex interplay between vaccination and the eye, focusing on the molecular and immunological pathways implicated in vaccine-associated ocular adverse effects. Through an in-depth analysis of recent advancements and the existing literature, we explored various mechanisms of vaccine-induced ocular inflammation, such as direct infection by live attenuated vaccines, immune complex formation, adjuvant-induced autoimmunity, molecular mimicry, hypersensitivity reactions, PEG-induced allergic reactions, Type 1 IFN activation, free extracellular RNA, and specific components. We further examined the specific ocular conditions associated with vaccination, such as uveitis, optic neuritis, and retinitis, and discussed the potential impact of novel vaccines, including those against SARS-CoV-2. This review sheds light on the intricate relationships between vaccination, the immune system, and ocular tissues, offering insights into informed discussions and future research directions aimed at optimizing vaccine safety and ophthalmological care. Our analysis underscores the importance of vigilance and further research to understand and mitigate the ocular side effects of vaccines, thereby ensuring the continued success of vaccination programs, while preserving ocular health.
Collapse
Affiliation(s)
| | - Koju Kamoi
- Department of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan; (Y.Z.); (Y.Z.); (J.Z.); (M.Y.); (K.O.-M.)
| | | | | | | | | |
Collapse
|
33
|
Drinković N, Beus M, Barbir R, Debeljak Ž, Tariba Lovaković B, Kalčec N, Ćurlin M, Bekavac A, Gorup D, Mamić I, Mandić D, Micek V, Turčić P, Günday-Türeli N, Türeli E, Vinković Vrček I. Novel PLGA-based nanoformulation decreases doxorubicin-induced cardiotoxicity. NANOSCALE 2024. [PMID: 38650478 DOI: 10.1039/d3nr06269d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Nanotechnology has the potential to provide formulations of antitumor agents with increased selectivity towards cancer tissue thereby decreasing systemic toxicity. This in vivo study evaluated the potential of novel nanoformulation based on poly(lactic-co-glycolic acid) (PLGA) to reduce the cardiotoxic potential of doxorubicin (DOX). In vivo toxicity of PLGADOX was compared with clinically approved non-PEGylated, liposomal nanoformulation of DOX (LipoDOX) and conventional DOX form (ConvDOX). The study was performed using Wistar Han rats of both sexes that were treated intravenously for 28 days with 5 doses of tested substances at intervals of 5 days. Histopathological analyses of heart tissues showed the presence of myofiber necrosis, degeneration processes, myocytolysis, and hemorrhage after treatment with ConvDOX, whereas only myofiber degeneration and hemorrhage were present after the treatment with nanoformulations. All DOX formulations caused an increase in the troponin T with the greatest increase caused by convDOX. qPCR analyses revealed an increase in the expression of inflammatory markers IL-6 and IL-8 after ConvDOX and an increase in IL-8 expression after lipoDOX treatments. The mass spectra imaging (MSI) of heart tissue indicates numerous metabolic and lipidomic changes caused by ConvDOX, while less severe cardiac damages were found after treatment with nanoformulations. In the case of LipoDOX, autophagy and apoptosis were still detectable, whereas PLGADOX induced only detectable mitochondrial toxicity. Cardiotoxic effects were frequently sex-related with the greater risk of cardiotoxicity observed mostly in male rats.
Collapse
Affiliation(s)
| | - Maja Beus
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Rinea Barbir
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Željko Debeljak
- JJ Strossmayer University of Osijek, Faculty of Medicine, Osijek, Croatia
- University Hospital Osijek, Osijek, Croatia
| | | | - Nikolina Kalčec
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | | | - Ana Bekavac
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Dunja Gorup
- Department of Neuroradiology, Klinik für Neuroradiology, Universitätspital Zürich Universitätsspital Zürich, 8006 Zürich, Switzerland
| | - Ivan Mamić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | | | - Vedran Micek
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| | - Petra Turčić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | | | | | - Ivana Vinković Vrček
- Institute for Medical Research and Occupational Health, Zagreb, Croatia.
- University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
34
|
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
|
35
|
Bitounis D, Jacquinet E, Rogers MA, Amiji MM. Strategies to reduce the risks of mRNA drug and vaccine toxicity. Nat Rev Drug Discov 2024; 23:281-300. [PMID: 38263456 DOI: 10.1038/s41573-023-00859-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/25/2024]
Abstract
mRNA formulated with lipid nanoparticles is a transformative technology that has enabled the rapid development and administration of billions of coronavirus disease 2019 (COVID-19) vaccine doses worldwide. However, avoiding unacceptable toxicity with mRNA drugs and vaccines presents challenges. Lipid nanoparticle structural components, production methods, route of administration and proteins produced from complexed mRNAs all present toxicity concerns. Here, we discuss these concerns, specifically how cell tropism and tissue distribution of mRNA and lipid nanoparticles can lead to toxicity, and their possible reactogenicity. We focus on adverse events from mRNA applications for protein replacement and gene editing therapies as well as vaccines, tracing common biochemical and cellular pathways. The potential and limitations of existing models and tools used to screen for on-target efficacy and de-risk off-target toxicity, including in vivo and next-generation in vitro models, are also discussed.
Collapse
Affiliation(s)
- Dimitrios Bitounis
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Moderna, Inc., Cambridge, MA, USA
| | | | | | - Mansoor M Amiji
- Departments of Pharmaceutical Sciences and Chemical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
36
|
Zhou J, Gao B, Zhang H, Yang R, Huang J, Li X, Zhong Y, Wang Y, Zhu X, Luo Y, Yan F. Ginsenoside modified lipid-coated perfluorocarbon nanodroplets: A novel approach to reduce complement protein adsorption and prolong in vivo circulation. Acta Pharm Sin B 2024; 14:1845-1863. [PMID: 38572112 PMCID: PMC10985128 DOI: 10.1016/j.apsb.2023.11.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 04/05/2024] Open
Abstract
Lipid-coated perfluorocarbon nanodroplets (lp-NDs) hold great promise in bio-medicine as vehicles for drug delivery, molecular imaging and vaccine agents. However, their clinical utility is restricted by limited targeted accumulation, attributed to the innate immune system (IIS), which acts as the initial defense mechanism in humans. This study aimed to optimize lp-ND formulations to minimize non-specific clearance by the IIS. Ginsenosides (Gs), the principal components of Panax ginseng, possessing complement inhibition ability, structural similarity to cholesterol, and comparable fat solubility to phospholipids, were used as promising candidate IIS inhibitors. Two different types of ginsenoside-based lp-NDs (Gs lp-NDs) were created, and their efficacy in reducing IIS recognition was examined. The Gs lp-NDs were observed to inhibit the adsorption of C3 in the protein corona (PC) and the generation of SC5b-9. Adding Gs to lp-NDs reduced complement adsorption and phagocytosis, resulting in a longer blood circulation time in vivo compared to lp-NDs that did not contain Gs. These results suggest that Gs can act as anti-complement and anti-phagocytosis adjuvants, potentially reducing non-specific clearance by the IIS and improving lifespan.
Collapse
Affiliation(s)
- Jie Zhou
- Ultrasound Department of West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Ultrasound Imaging of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Binyang Gao
- Ultrasound Department of West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Ultrasound Imaging of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huan Zhang
- Ultrasound Department of West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Ultrasound Imaging of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rui Yang
- Ultrasound Department of West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Ultrasound Imaging of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianbo Huang
- Ultrasound Department of West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Ultrasound Imaging of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Li
- West China Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Zhong
- West China Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Wang
- Research Core Facilities of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoxia Zhu
- Ultrasound Department of West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Ultrasound Imaging of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Luo
- Ultrasound Department of West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Ultrasound Imaging of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Yan
- Ultrasound Department of West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Ultrasound Imaging of West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
37
|
Rattan A, Malemnganba T, Sagar, Prajapati VK. Exploring structural engineering approach to formulate and characterize next-generation adjuvants. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:59-90. [PMID: 38762280 DOI: 10.1016/bs.apcsb.2023.12.020] [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: 05/20/2024]
Abstract
It is critical to emphasize the importance of vaccination as it protects us against harmful pathogens. Despite significant progress in vaccine development, there is an ongoing need to develop vaccines that are not only safe but also highly effective in protecting against severe infections. Subunit vaccines are generally safe, but they frequently fail to elicit strong immune responses. As a result, there is a need to improve vaccine effectiveness by combining them with adjuvants, which have the potential to boost the immune system many folds. The process of developing these adjuvants requires searching for molecules capable of activating the immune system, combining these promising compounds with an antigen, and then testing this combination using animal models before approving it for clinical use. Liposomal adjuvants work as delivery adjuvants and its activity depends on certain parameters such as surface charge, vesicle size, surface modification and route of administration. Self-assembly property of peptide adjuvants and discovery of hybrid peptides have widened the scope of peptides in vaccine formulations. Since most pathogenic molecules are not peptide based, phage display technique allows for screening peptide mimics for such pathogens that have potential as adjuvants. This chapter discusses about peptide and liposome-based adjuvants focusing on their properties imparting adjuvanticity along with the methods of formulating them. Methods of adjuvant characterization important for an adjuvant to be approved for clinical trials are also discussed. These include assays for cytotoxicity, T-lymphocyte proliferation, dendritic cell maturation, cytokine and antibody production, toll-like receptor dependent signaling and adjuvant half-life.
Collapse
Affiliation(s)
- Aditi Rattan
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Takhellambam Malemnganba
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sagar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
38
|
Estapé Senti M, García Del Valle L, Schiffelers RM. mRNA delivery systems for cancer immunotherapy: Lipid nanoparticles and beyond. Adv Drug Deliv Rev 2024; 206:115190. [PMID: 38307296 DOI: 10.1016/j.addr.2024.115190] [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: 10/31/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
mRNA-based vaccines are emerging as a promising alternative to standard cancer treatments and the conventional vaccines. Moreover, the FDA-approval of three nucleic acid based therapeutics (Onpattro, BNT162b2 and mRNA-1273) has further increased the interest and trust on this type of therapeutics. In order to achieve a significant therapeutic efficacy, the mRNA needs from a drug delivery system. In the last years, several delivery platforms have been explored, being the lipid nanoparticles (LNPs) the most well characterized and studied. A better understanding on how mRNA-based therapeutics operate (both the mRNA itself and the drug delivery system) will help to further improve their efficacy and safety. In this review, we will provide an overview of what mRNA cancer vaccines are and their mode of action and we will highlight the advantages and challenges of the different delivery platforms that are under investigation.
Collapse
Affiliation(s)
- Mariona Estapé Senti
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Lucía García Del Valle
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Raymond M Schiffelers
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
39
|
Yu YF, Wu EC, Lin SQ, Chu YX, Yang Y, Pan F, Ding TH, Qian J, Jiang K, Zhan CY. Reexamining the effects of drug loading on the in vivo performance of PEGylated liposomal doxorubicin. Acta Pharmacol Sin 2024; 45:646-659. [PMID: 37845342 PMCID: PMC10834505 DOI: 10.1038/s41401-023-01169-5] [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: 06/29/2023] [Accepted: 09/13/2023] [Indexed: 10/18/2023] Open
Abstract
Higher drug loading employed in nanoscale delivery platforms is a goal that researchers have long sought after. But such viewpoint remains controversial because the impacts that nanocarriers bring about on bodies have been seriously overlooked. In the present study we investigated the effects of drug loading on the in vivo performance of PEGylated liposomal doxorubicin (PLD). We prepared PLDs with two different drug loading rates: high drug loading rate, H-Dox, 12.9% w/w Dox/HSPC; low drug loading rate, L-Dox, 2.4% w/w Dox/HSPC (L-Dox had about 5 folds drug carriers of H-Dox at the same Dox dose). The pharmaceutical properties and biological effects of H-Dox and L-Dox were compared in mice, rats or 4T1 subcutaneous tumor-bearing mice. We showed that the lowering of doxorubicin loading did not cause substantial shifts to the pharmaceutical properties of PLDs such as in vitro and in vivo stability (stable), anti-tumor effect (equivalent effective), as well as tissue and cellular distribution. Moreover, it was even more beneficial for mitigating the undesired biological effects caused by PLDs, through prolonging blood circulation and alleviating cutaneous accumulation in the presence of pre-existing anti-PEG Abs due to less opsonins (e.g. IgM and C3) deposition on per particle. Our results warn that the effects of drug loading would be much more convoluted than expected due to the complex intermediation between nanocarriers and bodies, urging independent investigation for each individual delivery platform to facilitate clinical translation and application.
Collapse
Affiliation(s)
- Yi-Fei Yu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Er-Can Wu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Shi-Qi Lin
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Yu-Xiu Chu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Yang Yang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Feng Pan
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China
| | - Tian-Hao Ding
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Jun Qian
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China.
| | - Kuan Jiang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China.
- Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Chang-You Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China.
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, 201203, China.
| |
Collapse
|
40
|
Lin YJ, Zimmermann J, Schülke S. Novel adjuvants in allergen-specific immunotherapy: where do we stand? Front Immunol 2024; 15:1348305. [PMID: 38464539 PMCID: PMC10920236 DOI: 10.3389/fimmu.2024.1348305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Type I hypersensitivity, or so-called type I allergy, is caused by Th2-mediated immune responses directed against otherwise harmless environmental antigens. Currently, allergen-specific immunotherapy (AIT) is the only disease-modifying treatment with the potential to re-establish clinical tolerance towards the corresponding allergen(s). However, conventional AIT has certain drawbacks, including long treatment durations, the risk of inducing allergic side effects, and the fact that allergens by themselves have a rather low immunogenicity. To improve AIT, adjuvants can be a powerful tool not only to increase the immunogenicity of co-applied allergens but also to induce the desired immune activation, such as promoting allergen-specific Th1- or regulatory responses. This review summarizes the knowledge on adjuvants currently approved for use in human AIT: aluminum hydroxide, calcium phosphate, microcrystalline tyrosine, and MPLA, as well as novel adjuvants that have been studied in recent years: oil-in-water emulsions, virus-like particles, viral components, carbohydrate-based adjuvants (QS-21, glucans, and mannan) and TLR-ligands (flagellin and CpG-ODN). The investigated adjuvants show distinct properties, such as prolonging allergen release at the injection site, inducing allergen-specific IgG production while also reducing IgE levels, as well as promoting differentiation and activation of different immune cells. In the future, better understanding of the immunological mechanisms underlying the effects of these adjuvants in clinical settings may help us to improve AIT.
Collapse
Affiliation(s)
- Yen-Ju Lin
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
- Section Research Allergology (ALG 5), Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
41
|
Lu B, Lim JM, Yu B, Song S, Neeli P, Sobhani N, K P, Bonam SR, Kurapati R, Zheng J, Chai D. The next-generation DNA vaccine platforms and delivery systems: advances, challenges and prospects. Front Immunol 2024; 15:1332939. [PMID: 38361919 PMCID: PMC10867258 DOI: 10.3389/fimmu.2024.1332939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024] Open
Abstract
Vaccines have proven effective in the treatment and prevention of numerous diseases. However, traditional attenuated and inactivated vaccines suffer from certain drawbacks such as complex preparation, limited efficacy, potential risks and others. These limitations restrict their widespread use, especially in the face of an increasingly diverse range of diseases. With the ongoing advancements in genetic engineering vaccines, DNA vaccines have emerged as a highly promising approach in the treatment of both genetic diseases and acquired diseases. While several DNA vaccines have demonstrated substantial success in animal models of diseases, certain challenges need to be addressed before application in human subjects. The primary obstacle lies in the absence of an optimal delivery system, which significantly hampers the immunogenicity of DNA vaccines. We conduct a comprehensive analysis of the current status and limitations of DNA vaccines by focusing on both viral and non-viral DNA delivery systems, as they play crucial roles in the exploration of novel DNA vaccines. We provide an evaluation of their strengths and weaknesses based on our critical assessment. Additionally, the review summarizes the most recent advancements and breakthroughs in pre-clinical and clinical studies, highlighting the need for further clinical trials in this rapidly evolving field.
Collapse
Affiliation(s)
- Bowen Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Ming Lim
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Boyue Yu
- Department of Environmental Science, Policy, and Management, University of California at Berkeley, Berkeley, CA, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Pavithra K
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
42
|
Yin F, Xu X, Qi J, Guo M, Wang Y, Wang Y, Ye R, Lin Q, Yang D, Zhu X, Wang J. DSPE-PEG 2000-methotrexate nanoparticles encapsulating phenobarbital sodium kill cancer cells by inducing pyroptosis. J Mol Med (Berl) 2024; 102:213-229. [PMID: 38047923 DOI: 10.1007/s00109-023-02403-7] [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: 05/27/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/05/2023]
Abstract
Cancer is a life-threatening disease worldwide. Nanomedicine and nanodelivery systems are recently developed scientific field that employs specific materials in the nanoscale range to deliver drugs. Lipid-based nanoparticles are an ideal delivery system since they exhibit many advantages, including high bioavailability, self-assembly, formulation simplicity, and the ability to exhibit a plethora of physicochemical properties. Herein, we report that phenobarbital sodium can kill cancer cells by using the DSPE-PEG2000-methotrexate nanoparticle delivery system, which can target folate receptors that are usually overexpressed on a variety of cancer cells. The released phenobarbital then executes cancer cells by inducing pyroptosis. Results from our animal model further indicate that the nanomedicine of nanoparticle-encapsulated phenobarbital sodium is a promising anticancer therapy.
Collapse
Affiliation(s)
- Fengyue Yin
- Department of Emergency, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361005, China
- Department of Pharmacy, Medical College of Guangxi University, Nanning, 530004, Guangxi, China
| | - Xiao Xu
- Department of Pharmacy, Medical College of Guangxi University, Nanning, 530004, Guangxi, China
| | - Julia Qi
- Peking University Health Science Center, Beijing, 100191, China
| | - Mengyu Guo
- Department of Emergency, Zhongshan Hospital of Xiamen University, Xiamen, 361005, Fujian, China
| | - Yubo Wang
- Department of Biomedical Engineering, Medical College of Guangxi University, Nanning, 530004, Guangxi, China
| | - Yun Wang
- Department of Internal Medicine, School of Clinical Medicine, Jiamusi University, Heilongjiang 154007, Jiamusi, China
| | - Roumei Ye
- Department of Pharmacy, Medical College of Guangxi University, Nanning, 530004, Guangxi, China
| | - Qian Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005, Fujian, China
| | - Daowei Yang
- Department of Clinical Sciences, Lund University, 21428, Malmö, Sweden.
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005, Fujian, China.
| | - Jinling Wang
- Department of Emergency, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
43
|
Yang M, Zhang Z, Jin P, Jiang K, Xu Y, Pan F, Tian K, Yuan Z, Liu XE, Fu J, Wang B, Yan H, Zhan C, Zhang Z. Effects of PEG antibodies on in vivo performance of LNP-mRNA vaccines. Int J Pharm 2024; 650:123695. [PMID: 38081560 DOI: 10.1016/j.ijpharm.2023.123695] [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: 09/07/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 12/22/2023]
Abstract
Polyethylene glycol (PEG) plays important roles in stabilizing and lengthening circulation time of lipid nanoparticle (LNP) vaccines. Nowadays various levels of PEG antibodies have been detected in human blood, but the impact and mechanism of PEG antibodies on the in vivo performance of LNP vaccines has not been clarified thoroughly. By illustrating the distribution characteristics of PEG antibodies in human, the present study focused on the influence of PEG antibodies on the safety and efficacy of LNP-mRNA vaccine against COVID-19 in animal models. It was found that PEG antibodies led to shortened blood circulation duration, elevated accumulation and mRNA expression in liver and spleen, enhanced expression in macrophage and dendritic cells, while without affecting the production of anti-Spike protein antibodies of COVID-19 LNP vaccine. Noteworthily, PEG antibodies binding on the LNP vaccine increased probability of complement activation in animal as well as in human serum and led to lethal side effect in large dosage via intravenous injection of mice. Our data suggested that PEG antibodies in human was a risky factor of LNP-based vaccines for biosafety concerns but not efficacy.
Collapse
Affiliation(s)
- Min Yang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China
| | - Zengyu Zhang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China
| | - Pengpeng Jin
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China; Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University, Shanghai 201399, PR China
| | - Kuan Jiang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China; Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai 200031, PR China
| | - Yifei Xu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 PR China
| | - Feng Pan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, PR China
| | - Kaisong Tian
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China
| | - Zhou Yuan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China
| | | | - Jiaru Fu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, PR China
| | - Bin Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, PR China
| | - Huafang Yan
- Department of Health Management, Pudong Hospital, Fudan University, Shanghai 201399, PR China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China; State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 PR China; Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai 200032, PR China.
| | - Zui Zhang
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
44
|
Sakai H, Kure T, Kobayashi N, Ito T, Yamada Y, Yamada T, Miyamoto R, Imaizumi T, Ando J, Soga T, Osanai Y, Ogawa M, Shimizu T, Ishida T, Azuma H. Absence of Anaphylactic Reactions to Injection of Hemoglobin Vesicles (Artificial Red Cells) to Rodents. ACS OMEGA 2024; 9:1904-1915. [PMID: 38222647 PMCID: PMC10785325 DOI: 10.1021/acsomega.3c08641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 01/16/2024]
Abstract
The safety and efficacy of hemoglobin vesicles (HbVs) as artificial oxygen carriers encapsulating a purified and concentrated Hb solution in liposomes have been studied extensively. The HbV surface, modified with PEG by incorporating a PEG-conjugated phospholipid, is beneficial for storage and biocompatibility. However, it might be possible that interaction of PEG and the pre-existing anti-PEG antibody in the bloodstream causes acute adverse reaction. This study used two sets of experiments with rats and guinea pigs to ascertain whether the anti-PEG antibody generated by the PEG-modified HbV injection can induce anaphylactic reactions. SD rats received repeated intravenous injection of HbV at a dose rate of 16 or 32 mL/kg three times. Not anti-PEG IgG but anti-PEG IgM was detected. Nevertheless, no anaphylactic reaction occurred. Guinea pigs were used to study the presence of active systemic anaphylaxis further after injections of the PEG-modified liposomes used for HbV. The animals were sensitized by three repeated subcutaneous injections of PEG-modified liposomes (PEG-liposome) along with adjuvant at 1 week intervals. For comparison, unmodified liposomes (liposome) and 10 times excessively PEG-modified liposomes with ionizable lipid (10PEG-DODAP-liposome) were used. Inclusion of PEG modification induced not only anti-PEG IgM but also anti-PEG IgG. Three weeks after the final injection, intravenous injection of both PEG-liposome and liposome (1 mL/kg) induced no anaphylactic reaction. However, the injection of 10PEG-DODAP-liposome showed one lethal anaphylaxis case and one mild anaphylaxis case. Antisera obtained from the animal sensitized as described above were inoculated (0.05 mL) intradermally into fresh guinea pigs. The presence of passive cutaneous anaphylaxis was evaluated after intravenous injections (1 mL/kg) of three liposomes with Evans blue. No dye leakage was detected at any inoculated skin point for PEG-liposome or liposome, but a slight leakage was detected in one inoculated skin point for 10PEG-DODAP-liposome. These results indicate the absence of acute allergic reactions at repeated injections of HbVs despite the anti-PEG antibody induction. Not all the PEG-modified liposomes show anaphylaxis, and it may depend on the amount of PEGylated phospholipid and lipid composition of PEG-modified liposomes.
Collapse
Affiliation(s)
- Hiromi Sakai
- Department
of Chemistry, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Tomoko Kure
- Department
of Chemistry, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Naoko Kobayashi
- Department
of Chemistry, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Tadashi Ito
- Nihon
Bioresearch Inc., Hashima 501-6251, Japan
| | | | | | | | | | - Jiro Ando
- Nihon
Bioresearch Inc., Hashima 501-6251, Japan
| | | | | | | | - Taro Shimizu
- Research
Institute for Microbial Diseases, Osaka
University, Suita 565-0871, Japan
- Department
of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical
Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Tatsuhiro Ishida
- Department
of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical
Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Hiroshi Azuma
- Department
of Pediatrics, Asahikawa Medical University, Asahikawa 078-8510, Japan
| |
Collapse
|
45
|
Marquez CA, Oh CI, Ahn G, Shin WR, Kim YH, Ahn JY. Synergistic vesicle-vector systems for targeted delivery. J Nanobiotechnology 2024; 22:6. [PMID: 38167116 PMCID: PMC10763086 DOI: 10.1186/s12951-023-02275-6] [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: 07/28/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
With the immense progress in drug delivery systems (DDS) and the rise of nanotechnology, challenges such as target specificity remain. The vesicle-vector system (VVS) is a delivery system that uses lipid-based vesicles as vectors for a targeted drug delivery. When modified with target-probing materials, these vesicles become powerful vectors for drug delivery with high target specificity. In this review, we discuss three general types of VVS based on different modification strategies: (1) vesicle-probes; (2) vesicle-vesicles; and (3) genetically engineered vesicles. The synthesis of each VVS type and their corresponding properties that are advantageous for targeted drug delivery, are also highlighted. The applications, challenges, and limitations of VVS are briefly examined. Finally, we share a number of insights and perspectives regarding the future of VVS as a targeted drug delivery system at the nanoscale.
Collapse
Affiliation(s)
- Christine Ardelle Marquez
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Cho-Im Oh
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Gna Ahn
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Woo-Ri Shin
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
- Department of Bioengineering, University of Pennsylvania, 210 S 33rd St, Philadelphia, PA, 19104, USA
| | - Yang-Hoon Kim
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Ji-Young Ahn
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
46
|
Fu P, Yin S, Cheng H, Xu W, Jiang J. Engineered Exosomes for Drug Delivery in Cancer Therapy: A Promising Approach and Application. Curr Drug Deliv 2024; 21:817-827. [PMID: 37438904 DOI: 10.2174/1567201820666230712103942] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 07/14/2023]
Abstract
A significant amount of research effort is currently focused on investigating the role of exosomes in various cancers. These tiny vesicles, apart from acting as biomarkers, also play a crucial role in tumor formation and development. Several studies have demonstrated that exosomes can be a drug delivery vehicle for cancer therapy. In this paper, we highlight the key advantages of exosomes as a drug delivery candidate, with a particular focus on their low immunogenicity, natural targeting ability and suitable mechanical properties. Furthermore, we propose that the selection of appropriate exosomes and drug loading methods based on therapeutic goals and product heterogeneity is essential for preparing engineered exosomes. We comprehensively analyzed the superiorities of current drug-loading methods to improve the creation of designed exosomes. Moreover, we systematically review the applications of engineered exosomes in various therapies such as immunotherapy, gene therapy, protein therapy, chemotherapy, indicating that engineered exosomes have the potential to be reliable and, safe drug carriers that can address the unmet needs in cancer clinical practice.
Collapse
Affiliation(s)
- Peiwen Fu
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, 215600, Jiangsu, China
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Siqi Yin
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Huiying Cheng
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, 215600, Jiangsu, China
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Wenrong Xu
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, 215600, Jiangsu, China
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jiajia Jiang
- Aoyang Cancer Institute, Affiliated Aoyang Hospital of Jiangsu University, Zhangjiagang, 215600, Jiangsu, China
- Jiangsu Province Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| |
Collapse
|
47
|
Jiang Y, Li W, Wang Z, Lu J. Lipid-Based Nanotechnology: Liposome. Pharmaceutics 2023; 16:34. [PMID: 38258045 PMCID: PMC10820119 DOI: 10.3390/pharmaceutics16010034] [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: 11/06/2023] [Revised: 12/18/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Over the past several decades, liposomes have been extensively developed and used for various clinical applications such as in pharmaceutical, cosmetic, and dietetic fields, due to its versatility, biocompatibility, and biodegradability, as well as the ability to enhance the therapeutic index of free drugs. However, some challenges remain unsolved, including liposome premature leakage, manufacturing irreproducibility, and limited translation success. This article reviews various aspects of liposomes, including its advantages, major compositions, and common preparation techniques, and discusses present U.S. FDA-approved, clinical, and preclinical liposomal nanotherapeutics for treating and preventing a variety of human diseases. In addition, we summarize the significance of and challenges in liposome-enabled nanotherapeutic development and hope it provides the fundamental knowledge and concepts about liposomes and their applications and contributions in contemporary pharmaceutical advancement.
Collapse
Affiliation(s)
- Yanhao Jiang
- Pharmaceutics and Pharmacokinetics Track, Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (Y.J.); (W.L.); (Z.W.)
| | - Wenpan Li
- Pharmaceutics and Pharmacokinetics Track, Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (Y.J.); (W.L.); (Z.W.)
| | - Zhiren Wang
- Pharmaceutics and Pharmacokinetics Track, Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (Y.J.); (W.L.); (Z.W.)
| | - Jianqin Lu
- Pharmaceutics and Pharmacokinetics Track, Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (Y.J.); (W.L.); (Z.W.)
- Clinical and Translational Oncology Program, NCI-Designated University of Arizona Comprehensive Cancer Center, Tucson, AZ 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
48
|
Mondal S, Ghosh S. Liposome-Mediated Anti-Viral Drug Delivery Across Blood-Brain Barrier: Can Lipid Droplet Target Be Game Changers? Cell Mol Neurobiol 2023; 44:9. [PMID: 38123863 DOI: 10.1007/s10571-023-01443-4] [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: 07/18/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Lipid droplets (LDs) are subcellular organelles secreted from the endoplasmic reticulum (ER) that play a major role in lipid homeostasis. Recent research elucidates additional roles of LDs in cellular bioenergetics and innate immunity. LDs activate signaling cascades for interferon response and secretion of pro-inflammatory cytokines. Since balanced lipid homeostasis is critical for neuronal health, LDs play a crucial role in neurodegenerative diseases. RNA viruses enhance the secretion of LDs to support various phases of their life cycle in neurons which further leads to neurodegeneration. Targeting the excess LD formation in the brain could give us a new arsenal of antiviral therapeutics against neuroviruses. Liposomes are a suitable drug delivery system that could be used for drug delivery in the brain by crossing the Blood-Brain Barrier. Utilizing this, various pharmacological inhibitors and non-coding RNAs can be delivered that could inhibit the biogenesis of LDs or reduce their sizes, reversing the excess lipid-related imbalance in neurons. Liposome-Mediated Antiviral Drug Delivery Across Blood-Brain Barrier. Developing effective antiviral drug is challenging and it doubles against neuroviruses that needs delivery across the Blood-Brain Barrier (BBB). Lipid Droplets (LDs) are interesting targets for developing antivirals, hence targeting LD formation by drugs delivered using Liposomes can be game changers.
Collapse
Affiliation(s)
- Sourav Mondal
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Sourish Ghosh
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
49
|
Khunsuk PO, Pongma C, Palaga T, Hoven VP. Zwitterionic Polymer-Decorated Lipid Nanoparticles for mRNA Delivery in Mammalian Cells. Biomacromolecules 2023; 24:5654-5665. [PMID: 37956106 DOI: 10.1021/acs.biomac.3c00649] [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: 11/15/2023]
Abstract
Lipid nanoparticles (LNPs) play a key role in the effective transport of mRNA into cells for protein translation. Despite the stealthiness of poly(ethylene glycol) (PEG) that helps protect LNPs from protein absorption and blood clearance, the generation of anti-PEG antibodies resulting in PEG allergies remains a challenge for the development of an mRNA vaccine. Herein, a non-PEG lipid was developed by conjugating 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE) with an antifouling zwitterionic polymer, poly(2-methyacryloyloxyethyl phosphorylcholine) (PMPC), of different chain lengths. The PMPC-LNPs formulated from DPPE-PMPC were spherical (diameter ≈ 144-255 nm), neutral in charge, and stable at 4 °C for up to 28 days. Their fraction of stealthiness being close to 1 emphasized the antifouling characteristics of PMPC decorated on LNPs. The PMPC-LNPs were nontoxic to HEK293T cells, did not induce inflammatory responses in THP-1 cells, and exhibited an mRNA transfection efficiency superior to that of PEG-LNPs. This work demonstrated the potential of the developed zwitterionic polymer-conjugated LNPs as promising mRNA carriers.
Collapse
Affiliation(s)
- Phim-On Khunsuk
- Program in Petrochemistry and Polymer Science, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Chitsuda Pongma
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
- Center of Excellence in Materials and Bio-interfaces, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Voravee P Hoven
- Center of Excellence in Materials and Bio-interfaces, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
50
|
Maciel e Silva AT, Maia ALC, Silva JDO, Miranda SEM, Cantini TS, de Barros ALB, Soares DCF, de Magalhães MTQ, Alves RJ, Ramaldes GA. In Vitro and Preclinical Antitumor Evaluation of Doxorubicin Liposomes Coated with a Cholesterol-Based Trimeric β-D-Glucopyranosyltriazole. Pharmaceutics 2023; 15:2751. [PMID: 38140092 PMCID: PMC10747952 DOI: 10.3390/pharmaceutics15122751] [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: 11/14/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The coating of liposomes with polyethyleneglycol (PEG) has been extensively discussed over the years as a strategy for enhancing the in vivo and in vitro stability of nanostructures, including doxorubicin-loaded liposomes. However, studies have shown some important disadvantages of the PEG molecule as a long-circulation agent, including the immunogenic role of PEG, which limits its clinical use in repeated doses. In this context, hydrophilic molecules as carbohydrates have been proposed as an alternative to coating liposomes. Thus, this work studied the cytotoxicity and preclinical antitumor activity of liposomes coated with a glycosyl triazole glucose (GlcL-DOX) derivative as a potential strategy against breast cancer. The glucose-coating of liposomes enhanced the storage stability compared to PEG-coated liposomes, with the suitable retention of DOX encapsulation. The antitumor activity, using a 4T1 breast cancer mouse model, shows that GlcL-DOX controlled the tumor growth in 58.5% versus 35.3% for PEG-coated liposomes (PegL-DOX). Additionally, in the preliminary analysis of the GlcL-DOX systemic toxicity, the glucose-coating liposomes reduced the body weight loss and hepatotoxicity compared to other DOX-treated groups. Therefore, GlcL-DOX could be a promising alternative for treating breast tumors. Further studies are required to elucidate the complete GlcL-DOX safety profile.
Collapse
Affiliation(s)
- Aline Teixeira Maciel e Silva
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Ana Luiza Chaves Maia
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Juliana de Oliveira Silva
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Sued Eustáquio Mendes Miranda
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Talia Silva Cantini
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Andre Luis Branco de Barros
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Daniel Crístian Ferreira Soares
- Laboratório de Bioengenharia, Universidade Federal de Itajubá, Rua Irmã Ivone Drumond, 200, Distrito Industrial II, Itabira 35903-087, MG, Brazil;
| | - Mariana Torquato Quezado de Magalhães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil;
| | - Ricardo José Alves
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Gilson Andrade Ramaldes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| |
Collapse
|