1
|
Du Y, Tang C, Li B, Yue L, Su R, Qi W, Cui X, Wang Y. Role of Peptide Molecular Conformation in Improving the Gene Delivery Efficiency of Liposomal Formulations. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025. [PMID: 40397808 DOI: 10.1021/acs.langmuir.5c00351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
The study explored the impact of peptide conformations on the efficiency of nucleic acids transfection by designing two sets of linear (liKK and liRR) and cyclic peptides (cyKK and cyRR) with various sequences (KK: KKKKCRGDSCKKKK and RR: RRRRCLLLLCRRRR). Incorporating peptides in the liposome formulation improved gene transfection efficiency compared to the peptide and 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) alone. The results revealed that adding linear peptides demonstrated a higher delivery efficiency in comparison to cyclic peptides in transfection formulations. Additionally, two effective groups of transfection formulations were established. The liKK/DOTAP/DOPE formulation proved exceptional at delivering mRNA, achieving a transfection efficiency of 93.3%, comparable to that of Lipofectamine 2000 (91.7%). In contrast, the liRR/DOTAP/DOPE formulation effectively delivered both pDNA and siRNA. The pDNA transfection efficiency reached 42.3%, closely matching Lipofectamine 2000 (43.9%), while the siRNA transfection efficiency hit 90.7%, surpassing Lipofectamine 2000's performance (85.4%). The transfection efficiency of linear conformational formulations is superior to that of cyclic ones. Our research provides new options for liposome formulations and offers valuable insights for designing peptide-based gene delivery vectors with different conformations.
Collapse
Affiliation(s)
- Yaohui Du
- State Key Laboratory of Chemical Engineering and Low-Carbon Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Chuanmei Tang
- State Key Laboratory of Chemical Engineering and Low-Carbon Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Bowen Li
- State Key Laboratory of Chemical Engineering and Low-Carbon Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Lei Yue
- State Key Laboratory of Chemical Engineering and Low-Carbon Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Rongxin Su
- State Key Laboratory of Chemical Engineering and Low-Carbon Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072 P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Wei Qi
- State Key Laboratory of Chemical Engineering and Low-Carbon Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072 P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Xianbao Cui
- State Key Laboratory of Chemical Engineering and Low-Carbon Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Yuefei Wang
- State Key Laboratory of Chemical Engineering and Low-Carbon Technology, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
2
|
Kim J, Yang J, Heo S, Poo H. Evaluation of mRNA Transfection Reagents for mRNA Delivery and Vaccine Efficacy via Intramuscular Injection in Mice. ACS APPLIED BIO MATERIALS 2025; 8:4315-4324. [PMID: 40263125 DOI: 10.1021/acsabm.5c00424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The selection of an effective delivery carrier is crucial to assessing mRNA-based vaccines and therapeutics in vivo. Although lipid nanoparticles (LNPs) are commonly used for mRNA delivery, the LNP-mRNA formulation process is laborious and time-consuming and requires a high-cost microfluidic device. Instead, mixing with commercial reagents may simplify mRNA transfection into cells. However, their potential as in vivo carriers in intramuscular vaccination in mouse models remains unclear. In this study, we used three types of commercial RNA transfection reagents, MessengerMAX (MAX; liposome), TransIT-mRNA (IT; cationic polymer), and Invivofectamine (IVF; LNP), to produce nanoparticles directly by pipetting. The particle characteristics and mRNA delivery efficacy of the mRNA-transfection reagent mixtures were analyzed. Additionally, immune responses to vaccine efficacy and protective immunity of the mRNA mixtures as vaccine antigens were evaluated in a mouse model. Although MAX and IT showed high in vitro transfection efficiencies, their in vivo performances were limited. In contrast, IVF exhibited notable particle stability and homogeneity, making it a promising delivery carrier. Intramuscular IVF injection significantly enhanced both innate and adaptive immune responses with a robust systemic protein expression. Notably, when using SARS-CoV-2 Spike mRNA, IVF showed robust humoral immune responses, including production of IgG and neutralizing antibodies, thereby resulting in complete protection against SARS-CoV-2 infection. Therefore, these findings position IVF as an accessible and efficient mRNA carrier for evaluating mRNA vaccines and therapeutic efficacy in basic research.
Collapse
Affiliation(s)
- Jungho Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Jihyun Yang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Suhyeon Heo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Haryoung Poo
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
3
|
Talesh AA, Amiri S, Radi M, Hosseinifarahi M. Effect of nanocomposite alginate-based edible coatings containing thymol-nanoemulsion and/or thymol-loaded nanostructured lipid carriers on the microbial and physicochemical properties of carrot. Int J Biol Macromol 2025; 308:129196. [PMID: 38184040 DOI: 10.1016/j.ijbiomac.2023.129196] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/15/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
In this study, the effect of thymol-nanoemulsion (NE) and thymol-loaded nanostructured lipid carriers (NLC) on the physiological and microbial quality of carrot was investigated. The NE and NLC droplet sizes were 86 and 140 nm with encapsulation efficiency of 97 and 94 %, respectively. The minimum inhibitory concentration and minimum bactericidal concentration of thymol decreased in NE and increased in NLC against E. coli and S. aureus. Moreover, thymol-containing coatings exhibited a higher peroxidase activity, total phenolic content, flavonoid content, DPPH radical scavenging activity, pH, and lower respiration rate, TSS, weight loss, and decay with the preference for samples coated with NLC and NE (particularly NLC). The NE and NLC treatments significantly reduced the total viable, mold and yeast, lactic acid bacteria, and Enterobacteriaceae counts compared to the free thymol-containing coating. Results showed that the application of NE and NLC containing alginate-based coating (with the preference for NLC) improved the postharvest quality of carrot and extended its shelf life. Meanwhile, the separate application of these systems gave better results than the simultaneous application of both systems in one sample.
Collapse
Affiliation(s)
- Alireza Amiri Talesh
- Department of Food Science and Technology, Yasuj Branch, Islamic Azad University, Yasuj, Iran
| | - Sedigheh Amiri
- Department of Food Science and Technology, Yasuj Branch, Islamic Azad University, Yasuj, Iran; Sustainable Agriculture and Food Security Research Group, Yasuj Branch, Islamic Azad University, Yasuj, Iran.
| | - Mohsen Radi
- Department of Food Science and Technology, Yasuj Branch, Islamic Azad University, Yasuj, Iran; Sustainable Agriculture and Food Security Research Group, Yasuj Branch, Islamic Azad University, Yasuj, Iran.
| | - Mehdi Hosseinifarahi
- Sustainable Agriculture and Food Security Research Group, Yasuj Branch, Islamic Azad University, Yasuj, Iran; Department of Horticultural Science, Yasuj Branch, Islamic Azad University, Yasuj, Iran
| |
Collapse
|
4
|
Al-Shihabi AM, Al-Mohaya M, Haider M, Demiralp B. Exploring the promise of lipoplexes: From concept to clinical applications. Int J Pharm 2025; 674:125424. [PMID: 40043964 DOI: 10.1016/j.ijpharm.2025.125424] [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: 10/07/2024] [Revised: 02/02/2025] [Accepted: 02/28/2025] [Indexed: 03/09/2025]
Abstract
Lipoplexes are non-viral lipid vectors that effectively form complexes with genetic material, positioning them as promising alternatives to viral vectors in gene therapy. Their advantages include lower toxicity, reduced immunogenicity, improved targetability, and ease of large-scale production. A typical lipoplex is composed of cationic lipids, neutral lipids, and anionic nucleic acids (e.g., DNA, mRNA, miRNA, siRNA, shRNA). Neutral lipids play an auxiliary role and are often used as transfection enhancers. Enhancing lipoplex efficiency often involves modifying the cationic lipid structure through functional groups like PEG polymers and targeting ligands. The assembly of lipoplexes occurs spontaneously. This process involves the binding of the positively charged polar head group of the cationic lipid to the negatively charged DNA spontaneously as a result of electrostatic interaction, then irreversible rearrangement and condensation of the lipoplex occurs to form either lamellar or hexagonal structures. The transfection process encompasses several steps: cellular entry, endosomal escape and cargo release, cytoplasmic trafficking, and nuclear entry. The physicochemical and biological properties of lipoplexes are influenced by factors such as lipid structure, charge ratio, and environmental conditions. Despite certain limitations like low gene transfer efficiency and rapid clearance by serum proteins, lipoplexes show promise for clinical applications. They can be administered through various routes, offering potential treatments for diseases such as cancer, bone damage, infection, and cystic fibrosis. The study aims to examine the potential of lipoplexes as a promising vehicle for delivering therapeutic agents and their progression from theoretical concepts to practical clinical applications.
Collapse
Affiliation(s)
- Alaa M Al-Shihabi
- Institute of Health Sciences, Istanbul University, 34216, Beyazıt, Istanbul, Turkey; Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey
| | - Mazen Al-Mohaya
- Institute of Health Sciences, Istanbul University, 34216, Beyazıt, Istanbul, Turkey; Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey
| | - Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates.
| | - Burcu Demiralp
- Istanbul University, Faculty of Pharmacy, Pharmaceutical Technology Dept., 34126, Beyazıt, Istanbul, Turkey.
| |
Collapse
|
5
|
Kubbara EA, Bolad A, Malibary H. Advances in Liposomal Interleukin and Liposomal Interleukin Gene Therapy for Cancer: A Comprehensive Review of Preclinical Studies. Pharmaceutics 2025; 17:383. [PMID: 40143046 PMCID: PMC11945541 DOI: 10.3390/pharmaceutics17030383] [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: 12/13/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Preclinical studies on liposomal interleukin (IL) therapy demonstrate considerable promise in cancer treatment. This review explores the achievements, challenges, and future potential of liposomal IL encapsulation, focusing on preclinical studies. METHODS A structured search was conducted using the PubMed and Web of Science databases with the following search terms and Boolean operators: ("liposomal interleukin" OR "liposome-encapsulated interleukin") AND ("gene therapy" OR "gene delivery") AND ("cancer" OR "tumor" OR "oncology") AND ("pre-clinical studies" OR "animal models" OR "in vitro studies". RESULTS Liposomal IL-2 formulations are notable for enhancing delivery and retention at tumor sites. Recombinant human interleukin (rhIL-2) adsorbed onto small liposomes (35-50 nm) substantially reduces metastases in murine models. Hepatic metastasis models demonstrate superior efficacy of liposomal IL-2 over free IL-2 by enhancing immune responses, particularly in the liver. Localized delivery strategies, including nebulized liposomal IL-2 in canine pulmonary metastases and intrathoracic administration in murine sarcoma models, reduce systemic toxicity while promoting immune activation and tumor regression. Liposomal IL gene therapy, delivering cytokine genes directly to tumor sites, represents a notable advancement. Combining IL-2 gene therapy with other cytokines, including IL-6 or double-stranded RNA adjuvants, synergistically enhances macrophage and T-cell activation. Liposomal IL-4, IL-6, and IL-21 therapies show potential across various tumor types. Pairing liposomal IL-2 with chemotherapy or immune agents improves remission and survival. Innovative strategies, including PEGylation and ligand-targeted systems, optimize delivery, release, and therapeutic outcomes. CONCLUSIONS Utilizing immune-stimulatory ILs through advanced liposomal delivery and gene therapy establishes a strong foundation for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Eman A. Kubbara
- Clinical Biochemistry Department, Faculty of Medicine, Rabigh Branch, King Abdulaziz University, Rabigh 21911, Saudi Arabia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Ahmed Bolad
- Department of Microbiology and Unit of Immunology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Husam Malibary
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| |
Collapse
|
6
|
Paranjape S, Choi KY, Kashiwagi S, Choi HS. H-Dots: Renal Clearable Zwitterionic Nanocarriers for Disease Diagnosis and Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:2111-2123. [PMID: 39823416 DOI: 10.1021/acs.langmuir.4c04372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Nanocarriers have shown significant promise in the diagnosis and treatment of various diseases, utilizing a wide range of biocompatible materials such as metals, inorganic substances, and organic components. Despite diverse design strategies, key physicochemical properties, including hydrodynamic diameter, shape, surface charge, and hydrophilicity/lipophilicity, are crucial for optimizing biodistribution, pharmacokinetics, and therapeutic efficacy. However, these properties are often influenced by drug payload, presenting an ongoing challenge in developing versatile platform technologies for theranostics. To enable tissue- and organ-specific targeting while minimizing nonspecific uptake, renal clearable Harvard dots (H-dots) have emerged as a promising organic nanocarrier platform. Composed of an ε-polylysine backbone for a tunable charge, near-infrared fluorophores for tracking their fate in living organisms, and β-cyclodextrins for potential drug delivery, H-dots offer a multifunctional approach to theranostic nanomedicine. Recent studies demonstrate that H-dots are effective for targeted imaging and drug delivery to solid tumors. This review highlights current nanocarrier design strategies and recent advances in H-dot applications for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Swarali Paranjape
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Kyu Young Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
- Department of Otorhinolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital and Hallym University College of Medicine, Yeongdeungpo-gu, Seoul 07441, Republic of Korea
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
7
|
Tang L, Yang X, He L, Zhu C, Chen Q. Preclinical advance in nanoliposome-mediated photothermal therapy in liver cancer. Lipids Health Dis 2025; 24:31. [PMID: 39891269 PMCID: PMC11783920 DOI: 10.1186/s12944-024-02429-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/03/2025] Open
Abstract
Liver cancer is a highly lethal malignant tumor with a high incidence worldwide. Therefore, its treatment has long been a focus of medical research. Although traditional treatment methods such as surgery, radiotherapy, and chemotherapy have increased the survival rate of patients, their efficacy remains unsatisfactory owing to the nonspecific distribution of drugs, high toxicity, and drug resistance of tumor tissues. In recent years, the application of nanotechnology in the medical field has opened a new avenue for the treatment of liver cancer. Among these treatment methods, photothermal therapy (PTT) based on nanoliposomes has attracted wide attention owing to its unique targeting and high efficiency. This article reviews the latest preclinical research progress of nanoliposome-based PTT for liver cancer and its metastasis, discusses the preclinical challenges in this field, and proposes directions for improvement, with the aim of improving the effectiveness of liver cancer treatment.
Collapse
Affiliation(s)
- Lixuan Tang
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiao Yang
- The department of oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Liwen He
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chaogeng Zhu
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Qingshan Chen
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
8
|
Meinhard S, Erdmann F, Lucas H, Krabbes M, Krüger S, Wölk C, Mäder K. T14diLys/DOPE Liposomes: An Innovative Option for siRNA-Based Gene Knockdown? Pharmaceutics 2024; 17:25. [PMID: 39861674 PMCID: PMC11769127 DOI: 10.3390/pharmaceutics17010025] [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: 11/23/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Bringing small interfering RNA (siRNA) into the cell cytosol to achieve specific gene silencing is an attractive but also very challenging option for improved therapies. The first step for successful siRNA delivery is the complexation with a permanent cationic or ionizable compound. This protects the negatively charged siRNA and enables transfection through the cell membrane. The current study explores the performance of the innovative, ionizable lipid 2-Tetradecylhexadecanoic acid-(2-bis{[2-(2,6-diamino-1-oxohexyl)amino]ethyl}aminoethyl)-amide (T14diLys), in combination with 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), for siRNA delivery and the impact of the production method (sonication vs. extrusion) on the particle properties. METHODS Liposomes were produced either with sonication or extrusion and characterized. The extruded liposomes were combined with siRNA at different N/P ratios and investigated in terms of size zeta potential, encapsulation efficiency, lipoplex stability against RNase A, and knockdown efficiency using enhanced green fluorescent protein (eGFP)-marked colon adenocarcinoma cells. RESULTS The liposomes prepared by extrusion were smaller and had a narrower size distribution than the sonicated ones. The combination of siRNA and liposomes at a nitrogen-to-phosphate (N/P) ratio of 5 had optimal particle properties, high encapsulation efficiency, and lipoplex stability. Gene knockdown tests confirmed this assumption. CONCLUSIONS Liposomes produced with extrusion were more reproducible and provided enhanced particle properties. The physicochemical characterization and in vitro experiments showed that an N/P ratio of 5 was the most promising ratio for siRNA delivery.
Collapse
Affiliation(s)
- Sophie Meinhard
- Department of Pharmaceutical Technology, Faculty of Natural Sciences I, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle/Saale, Germany; (S.M.); (H.L.)
| | - Frank Erdmann
- Department of Pharmaceutical Pharmacology and Toxicology, Faculty of Natural Sciences I, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle/Saale, Germany;
- Research Center for Drug Therapy Halle, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany
| | - Henrike Lucas
- Department of Pharmaceutical Technology, Faculty of Natural Sciences I, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle/Saale, Germany; (S.M.); (H.L.)
- Research Center for Drug Therapy Halle, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany
| | - Maria Krabbes
- Pharmaceutical Technology, Medical Faculty, University of Leipzig, Eilenburger Strasse 15A, 04317 Leipzig, Germany; (M.K.); (C.W.)
| | - Stephanie Krüger
- Biocenter, Microscopy Unit, Martin Luther University Halle-Wittenberg, Weinbergweg 22, 06120 Halle/Saale, Germany;
| | - Christian Wölk
- Pharmaceutical Technology, Medical Faculty, University of Leipzig, Eilenburger Strasse 15A, 04317 Leipzig, Germany; (M.K.); (C.W.)
| | - Karsten Mäder
- Department of Pharmaceutical Technology, Faculty of Natural Sciences I, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle/Saale, Germany; (S.M.); (H.L.)
- Research Center for Drug Therapy Halle, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany
| |
Collapse
|
9
|
Pavlov RV, Akimov SA, Dashinimaev EB, Bashkirov PV. Boosting Lipofection Efficiency Through Enhanced Membrane Fusion Mechanisms. Int J Mol Sci 2024; 25:13540. [PMID: 39769303 PMCID: PMC11677079 DOI: 10.3390/ijms252413540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Gene transfection is a fundamental technique in the fields of biological research and therapeutic innovation. Due to their biocompatibility and membrane-mimetic properties, lipid vectors serve as essential tools in transfection. The successful delivery of genetic material into the cytoplasm is contingent upon the fusion of the vector and cellular membranes, which enables hydrophilic polynucleic acids to traverse the hydrophobic barriers of two intervening membranes. This review examines the critical role of membrane fusion in lipofection efficiency, with a particular focus on the molecular mechanisms that govern lipoplex-membrane interactions. This analysis will examine the key challenges inherent to the fusion process, from achieving initial membrane proximity to facilitating final content release through membrane remodeling. In contrast to viral vectors, which utilize specialized fusion proteins, lipid vectors necessitate a strategic formulation and environmental optimization to enhance their fusogenicity. This review discusses recent advances in vector design and fusion-promoting strategies, emphasizing their potential to improve gene delivery yield. It highlights the importance of understanding lipoplex-membrane fusion mechanisms for developing next-generation delivery systems and emphasizes the need for continued fundamental research to advance lipid-mediated transfection technology.
Collapse
Affiliation(s)
- Rais V. Pavlov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| | - Sergey A. Akimov
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, Moscow 119071, Russia;
| | - Erdem B. Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia;
| | - Pavel V. Bashkirov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| |
Collapse
|
10
|
Sarkar S, Moitra P, Bera S, Bhattacharya S. Antisense Oligonucleotide Embedded Context Responsive Nanoparticles Derived from Synthetic Ionizable Lipids for lncRNA Targeted Therapy of Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:45871-45887. [PMID: 39163516 DOI: 10.1021/acsami.4c04893] [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: 08/22/2024]
Abstract
The long noncoding RNAs (lncRNA) are primarily associated with several essential gene regulations but are also connected to cancer metabolism and progression. HOTAIR and MALAT1 are two such lncRNAs that are detected in malignancies of various origins and are responsible for the poor prognosis of cancer patients. Due to these factors, the lncRNAs have emerged as prime targets for the development of anticancer therapeutics. However, nonviral delivery of lncRNA-targeted antisense oligonucleotides (ASOs) still remains a critical challenge while maintaining their structural and functional integrity. Herein, we have designed and synthesized a new series of ionizable lipids with variations in their head groups to prepare lipid nanoparticle (LNP) formulation along with cholesterol-based twin cationic lipid and amphiphilic zwitterionic lipid. The context responsiveness of these formulations in delivering the ASOs has been thoroughly investigated by various bioanalytical techniques, and an optimum formulation has been identified. The LNPs are utilized to deliver the ASOs targeting HOTAIR lncRNA in human cancer cell lines and MALAT1 lncRNA in mouse models. This study thus standardizes an advanced nanomaterial system for nonviral gene delivery that has been validated by a considerable reduction in the target lncRNA level under in vitro and a significant reduction in tumor volume under in vivo settings.
Collapse
Affiliation(s)
- Sourav Sarkar
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Parikshit Moitra
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Berhampur, Odisha 760003, India
| | - Sayan Bera
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Santanu Bhattacharya
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
- Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata 700032, India
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, India
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati (IISER Tirupati), Srinivasapuram, Yerpedu Mandal, Tirupati District, Andhra Pradesh 517619, India
| |
Collapse
|
11
|
Fieni C, Sorrentino C, Ciummo SL, Fontana A, Lotti LV, Scialis S, Calvo Garcia D, Caulo M, Di Carlo E. Immunoliposome-based targeted delivery of the CRISPR/Cas9gRNA-IL30 complex inhibits prostate cancer and prolongs survival. Exp Mol Med 2024; 56:2033-2051. [PMID: 39232121 PMCID: PMC11447253 DOI: 10.1038/s12276-024-01310-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 09/06/2024] Open
Abstract
The development of selective and nontoxic immunotherapy targeting prostate cancer (PC) is challenging. Interleukin (IL)30 plays immunoinhibitory and oncogenic roles in PC, and its tumor-specific suppression may have significant clinical implications. CRISPR/Cas9-mediated IL30 gene deletion in PC xenografts using anti-PSCA antibody-driven lipid nanocomplexes (Cas9gRNA-hIL30-PSCA NxPs) revealed significant genome editing efficiency and circulation stability without off-target effects or organ toxicity. Biweekly intravenous administration of Cas9gRNA-hIL30-PSCA NxPs to PC-bearing mice inhibited tumor growth and metastasis and improved survival. Mechanistically, Cas9gRNA-hIL30-PSCA NxPs suppressed ANGPTL 1/2/4, IL1β, CCL2, CXCL1/6, SERPINE1-F1, EFNB2, PLG, PF4, VEGFA, VEGFD, ANG, TGFβ1, EGF and HGF expression in human PC cells while upregulated CDH1, DKK3 and PTEN expression, leading to low proliferation and extensive ischemic necrosis. In the syngeneic PC model, IL30-targeting immunoliposomes downregulated NFKB1 expression and prevented intratumoral influx of CD11b+Gr-1+MDCs, Foxp3+Tregs, and NKp46+RORγt+ILC3, and prolonged host survival by inhibiting tumor progression. This study serves as a proof of principle that immunoliposome-based targeted delivery of Cas9gRNA-IL30 represent a potentially safe and effective strategy for PC treatment.
Collapse
Affiliation(s)
- Cristiano Fieni
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Stefania Livia Ciummo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Antonella Fontana
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UDA-TECHLAB Research Center, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | | | - Sofia Scialis
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Darien Calvo Garcia
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Massimo Caulo
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
12
|
Barbieri BD, Peeler DJ, Samnuan K, Day S, Hu K, Sallah HJ, Tregoning JS, McKay PF, Shattock RJ. The role of helper lipids in optimising nanoparticle formulations of self-amplifying RNA. J Control Release 2024; 374:280-292. [PMID: 39142355 DOI: 10.1016/j.jconrel.2024.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Lipid nanoparticle (LNP) formulation plays a vital role in RNA vaccine delivery. However, further optimisation of self-amplifying RNA (saRNA) vaccine formulation could help enhance seroconversion rates in humans and improve storage stability. Altering either the ionisable or helper lipid can alter the characteristics and performance of formulated saRNA through the interplay of the phospholipid's packing parameter and the geometrical shape within the LNP membrane. In this study, we compared the impact of three helper lipids (DSPC, DOPC, or DOPE) used with two different ionisable lipids (MC3 and C12-200) on stability, transfection efficiency and the inflammation and immunogenicity of saRNA. While helper lipid identity altered saRNA expression across four cell lines in vitro, this was not predictive of an ex vivo or in vivo response. The helper lipid used influenced LNP storage where DSPC provided the best stability profile over four weeks at 2-8 °C. Importantly, helper lipid impact on LNP storage stability was the best predictor of expression in human skin explants, where C12-200 in combination with DSPC provided the most durable expression. C12-200 LNPs also improved protein expression (firefly luciferase) and humoral responses to a SARS-CoV-2 spike saRNA vaccine compared to MC3 LNPs, where the effect of helper lipids was less apparent. Nevertheless, the performance of C12-200 in combination with DSPC appears optimal for saRNA when balancing preferred storage stability requirements against in vivo and ex vivo potency. These data suggest that helper lipid influences the stability and functionality of ionisable lipid nanoparticle-formulated saRNA.
Collapse
Affiliation(s)
| | - David J Peeler
- Department of Infectious Disease, Imperial College London, London, UK; Department of Materials, Imperial College London, London, UK
| | - Karnyart Samnuan
- Department of Infectious Disease, Imperial College London, London, UK
| | - Suzanne Day
- Department of Infectious Disease, Imperial College London, London, UK
| | - Kai Hu
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, UK
| | - Paul F McKay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Robin J Shattock
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
13
|
Chan A, Haley RM, Najar MA, Gonzalez-Martinez D, Bugaj LJ, Burslem GM, Mitchell MJ, Tsourkas A. Lipid-mediated intracellular delivery of recombinant bioPROTACs for the rapid degradation of undruggable proteins. Nat Commun 2024; 15:5808. [PMID: 38987546 PMCID: PMC11237011 DOI: 10.1038/s41467-024-50235-x] [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: 11/02/2023] [Accepted: 07/04/2024] [Indexed: 07/12/2024] Open
Abstract
Recently, targeted degradation has emerged as a powerful therapeutic modality. Relying on "event-driven" pharmacology, proteolysis targeting chimeras (PROTACs) can degrade targets and are superior to conventional inhibitors against undruggable proteins. Unfortunately, PROTAC discovery is limited by warhead scarcity and laborious optimization campaigns. To address these shortcomings, analogous protein-based heterobifunctional degraders, known as bioPROTACs, have been developed. Compared to small-molecule PROTACs, bioPROTACs have higher success rates and are subject to fewer design constraints. However, the membrane impermeability of proteins severely restricts bioPROTAC deployment as a generalized therapeutic modality. Here, we present an engineered bioPROTAC template able to complex with cationic and ionizable lipids via electrostatic interactions for cytosolic delivery. When delivered by biocompatible lipid nanoparticles, these modified bioPROTACs can rapidly degrade intracellular proteins, exhibiting near-complete elimination (up to 95% clearance) of targets within hours of treatment. Our bioPROTAC format can degrade proteins localized to various subcellular compartments including the mitochondria, nucleus, cytosol, and membrane. Moreover, substrate specificity can be easily reprogrammed, allowing modular design and targeting of clinically-relevant proteins such as Ras, Jnk, and Erk. In summary, this work introduces an inexpensive, flexible, and scalable platform for efficient intracellular degradation of proteins that may elude chemical inhibition.
Collapse
Affiliation(s)
- Alexander Chan
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca M Haley
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohd Altaf Najar
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Gonzalez-Martinez
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Lukasz J Bugaj
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - George M Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Tsourkas
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Jin Y, Wang X, Kromer AP, Müller JT, Zimmermann C, Xu Z, Hartschuh A, Adams F, Merkel OM. Role of Hydrophobic Modification in Spermine-Based Poly(β-amino ester)s for siRNA Delivery and Their Spray-Dried Powders for Inhalation and Improved Storage. Biomacromolecules 2024; 25:4177-4191. [PMID: 38866384 PMCID: PMC11238323 DOI: 10.1021/acs.biomac.4c00283] [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: 02/28/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024]
Abstract
After RNAi was first discovered over 20 years ago, siRNA-based therapeutics are finally becoming reality. However, the delivery of siRNA has remained a challenge. In our previous research, we found that spermine-based poly(β-amino ester)s are very promising for siRNA delivery. However, the role of hydrophobic modification in siRNA delivery of spermine-based poly(β-amino ester)s is not fully understood yet. In the current work, we synthesized spermine-based poly(β-amino ester)s with different percentages of oleylamine side chains, named P(SpOABAE). The chemical structures of the polymers were characterized by 1H NMR. The polymers showed efficient siRNA encapsulation determined by SYBR Gold assays. The hydrodynamic diameters of the P(SpOABAE) polyplexes from charge ratio N/P 1 to 20 were 30-100 nm except for aggregation phenomena observed at N/P 3. Morphology of the polyplexes was visualized by atomic force microscopy, and cellular uptake was determined by flow cytometry in H1299 cells, where all the polyplexes showed significantly higher cellular uptake than hyperbranched polyethylenimine (25 kDa). The most hydrophobic P(SpOABAE) polyplexes were able to achieve more than 90% GFP knockdown in H1299/eGFP cells. The fact that gene silencing efficacy increased with hydrophobicity but cellular uptake was affected by both charge and hydrophobic interactions highlights the importance of endosomal escape. For pulmonary administration and improved storage stability, the polyplexes were spray-dried. Results confirmed the maintained siRNA activity after storage for 3 months at room temperature, indicating potential for dry powder inhalation.
Collapse
Affiliation(s)
- Yao Jin
- Department
of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Xiaoxuan Wang
- Department
of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Adrian P.E. Kromer
- Department
of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Joschka T. Müller
- Department
of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Christoph Zimmermann
- Department
of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Zehua Xu
- Department
of Chemistry, Ludwig-Maximilians-Universität
München, Butenandtstr.
5-13, 81377 Munich, Germany
| | - Achim Hartschuh
- Department
of Chemistry, Ludwig-Maximilians-Universität
München, Butenandtstr.
5-13, 81377 Munich, Germany
- Center
for Nanoscience (CeNS), Ludwig-Maximilians-Universität
München, 80799 München, Germany
| | - Friederike Adams
- Department
of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Olivia M. Merkel
- Department
of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany
- Center
for Nanoscience (CeNS), Ludwig-Maximilians-Universität
München, 80799 München, Germany
| |
Collapse
|
15
|
Duraloglu C, Baysal I, Yabanoglu-Ciftci S, Arica B. Nintedanib and miR-29b co-loaded lipoplexes in idiopathic pulmonary fibrosis: formulation, characterization, and in vitro evaluation. Drug Dev Ind Pharm 2024; 50:671-686. [PMID: 39099436 DOI: 10.1080/03639045.2024.2387166] [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: 03/21/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/06/2024]
Abstract
OBJECTIVE This study was aimed to develop a cationic lipoplex formulation loaded with Nintedanib and miR-29b (LP-NIN-miR) as an alternative approach in the combination therapy of idiopathic pulmonary dibrosis (IPF) by proving its additive anti-fibrotic therapeutic effects through in vitro lung fibrosis model. SIGNIFICANCE This is the first research article reported that the LP-NIN-MIR formulations in the treatment of IPF. METHODS To optimize cationic liposomes (LPs), quality by design (QbD) approach was carried out. Optimized blank LP formulation was prepared with DOTAP, CHOL, DOPE, and DSPE-mPEG 2000 at the molar ratio of 10:10:1:1. Nintedanib loaded LP (LPs-NIN) were produced by microfluidization method and were incubated with miR-29b at room temperature for 30 min to obtain LP-NIN-miR. To evaluate the cellular uptake of LP-NIN-miR, NIH/3T3 cells were treated with 20 ng.mL-1 transforming growth factor-β1 (TGF-β1) for 96 h to establish the in vitro IPF model and incubated with LP-NIN-miR for 48 h. RESULTS The hydrodynamic diameter, polydispersity index (PDI), and zeta potential of the LP-NIN-miR were 87.3 ± 0.9 nm, 0.184 ± 0.003, and +24 ± 1 mV, respectively. The encapsulation efficiencies of Nintedanib and miR-29b were 99.8% ± 0.08% and 99.7% ± 1.2%, respectively. The results of the cytotoxicity study conducted with NIH/3T3 cells indicated that LP-NIN-miR is a safe delivery system. CONCLUSIONS The outcome of the transfection study proved the additive anti-fibrotic therapeutic effect of LP-NIN-miR and suggested that lipoplexes are effective delivery systems for drug and nucleic acid to the NIH/3T3 cells in the treatment of IPF.
Collapse
Affiliation(s)
- Ceren Duraloglu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Ipek Baysal
- Vocational School of Health Services, Hacettepe University, Ankara, Turkey
| | | | - Betul Arica
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
16
|
Desai N, Rana D, Salave S, Benival D, Khunt D, Prajapati BG. Achieving Endo/Lysosomal Escape Using Smart Nanosystems for Efficient Cellular Delivery. Molecules 2024; 29:3131. [PMID: 38999083 PMCID: PMC11243486 DOI: 10.3390/molecules29133131] [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: 06/06/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
The delivery of therapeutic agents faces significant hurdles posed by the endo-lysosomal pathway, a bottleneck that hampers clinical effectiveness. This comprehensive review addresses the urgent need to enhance cellular delivery mechanisms to overcome these obstacles. It focuses on the potential of smart nanomaterials, delving into their unique characteristics and mechanisms in detail. Special attention is given to their ability to strategically evade endosomal entrapment, thereby enhancing therapeutic efficacy. The manuscript thoroughly examines assays crucial for understanding endosomal escape and cellular uptake dynamics. By analyzing various assessment methods, we offer nuanced insights into these investigative approaches' multifaceted aspects. We meticulously analyze the use of smart nanocarriers, exploring diverse mechanisms such as pore formation, proton sponge effects, membrane destabilization, photochemical disruption, and the strategic use of endosomal escape agents. Each mechanism's effectiveness and potential application in mitigating endosomal entrapment are scrutinized. This paper provides a critical overview of the current landscape, emphasizing the need for advanced delivery systems to navigate the complexities of cellular uptake. Importantly, it underscores the transformative role of smart nanomaterials in revolutionizing cellular delivery strategies, leading to a paradigm shift towards improved therapeutic outcomes.
Collapse
Affiliation(s)
- Nimeet Desai
- Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India;
| | - Dhwani Rana
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Dignesh Khunt
- School of Pharmacy, Gujarat Technological University, Gandhinagar 382027, Gujarat, India
| | - Bhupendra G. Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, Gujarat, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
17
|
El-Zahaby SA, Kaur L, Sharma A, Prasad AG, Wani AK, Singh R, Zakaria MY. Lipoplexes' Structure, Preparation, and Role in Managing Different Diseases. AAPS PharmSciTech 2024; 25:131. [PMID: 38849687 DOI: 10.1208/s12249-024-02850-6] [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: 02/18/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Lipid-based vectors are becoming promising alternatives to traditional therapies over the last 2 decades specially for managing life-threatening diseases like cancer. Cationic lipids are the most prevalent non-viral vectors utilized in gene delivery. The increasing number of clinical trials about lipoplex-based gene therapy demonstrates their potential as well-established technology that can provide robust gene transfection. In this regard, this review will summarize this important point. These vectors however have a modest transfection efficiency. This limitation can be partly addressed by using functional lipids that provide a plethora of options for investigating nucleic acid-lipid interactions as well as in vitro and in vivo nucleic acid delivery for biomedical applications. Despite their lower gene transfer efficiency, lipid-based vectors such as lipoplexes have several advantages over viral ones: they are less toxic and immunogenic, can be targeted, and are simple to produce on a large scale. Researchers are actively investigating the parameters that are essential for an effective lipoplex delivery method. These include factors that influence the structure, stability, internalization, and transfection of the lipoplex. Thorough understanding of the design principles will enable synthesis of customized lipoplex formulations for life-saving therapy.
Collapse
Affiliation(s)
- Sally A El-Zahaby
- Department of Pharmaceutics and Industrial Pharmacy, PharmD Program, Egypt-Japan University of Science and Technology (E-JUST), Alexandria, Egypt.
| | - Lovepreet Kaur
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, 144411, Punjab, India
| | - Ankur Sharma
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Aprameya Ganesh Prasad
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, 144411, Punjab, India
| | - Rattandeep Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, 144411, Punjab, India
| | - Mohamed Y Zakaria
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Port Said University, Port Said, 42526, Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, King Salman International University, Ras Sudr, 46612, South Sinai, Egypt
| |
Collapse
|
18
|
Fedorovskiy AG, Antropov DN, Dome AS, Puchkov PA, Makarova DM, Konopleva MV, Matveeva AM, Panova EA, Shmendel EV, Maslov MA, Dmitriev SE, Stepanov GA, Markov OV. Novel Efficient Lipid-Based Delivery Systems Enable a Delayed Uptake and Sustained Expression of mRNA in Human Cells and Mouse Tissues. Pharmaceutics 2024; 16:684. [PMID: 38794346 PMCID: PMC11125954 DOI: 10.3390/pharmaceutics16050684] [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: 04/18/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Over the past decade, mRNA-based therapy has displayed significant promise in a wide range of clinical applications. The most striking example of the leap in the development of mRNA technologies was the mass vaccination against COVID-19 during the pandemic. The emergence of large-scale technology and positive experience of mRNA immunization sparked the development of antiviral and anti-cancer mRNA vaccines as well as therapeutic mRNA agents for genetic and other diseases. To facilitate mRNA delivery, lipid nanoparticles (LNPs) have been successfully employed. However, the diverse use of mRNA therapeutic approaches requires the development of adaptable LNP delivery systems that can control the kinetics of mRNA uptake and expression in target cells. Here, we report effective mRNA delivery into cultured mammalian cells (HEK293T, HeLa, DC2.4) and living mouse muscle tissues by liposomes containing either 1,26-bis(cholest-5-en-3β-yloxycarbonylamino)-7,11,16,20-tetraazahexacosane tetrahydrochloride (2X3) or the newly applied 1,30-bis(cholest-5-en-3β-yloxycarbonylamino)-9,13,18,22-tetraaza-3,6,25,28-tetraoxatriacontane tetrahydrochloride (2X7) cationic lipids. Using end-point and real-time monitoring of Fluc mRNA expression, we showed that these LNPs exhibited an unusually delayed (of over 10 h in the case of the 2X7-based system) but had highly efficient and prolonged reporter activity in cells. Accordingly, both LNP formulations decorated with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (DSPE-PEG2000) provided efficient luciferase production in mice, peaking on day 3 after intramuscular injection. Notably, the bioluminescence was observed only at the site of injection in caudal thigh muscles, thereby demonstrating local expression of the model gene of interest. The developed mRNA delivery systems hold promise for prophylactic applications, where sustained synthesis of defensive proteins is required, and open doors to new possibilities in mRNA-based therapies.
Collapse
Affiliation(s)
- Artem G. Fedorovskiy
- Belozersky Institute of Physico-Chemical Biology, Department of Materials Science, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.G.F.); (M.V.K.); (E.A.P.)
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Denis N. Antropov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| | - Anton S. Dome
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| | - Pavel A. Puchkov
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Daria M. Makarova
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Maria V. Konopleva
- Belozersky Institute of Physico-Chemical Biology, Department of Materials Science, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.G.F.); (M.V.K.); (E.A.P.)
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Anastasiya M. Matveeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| | - Eugenia A. Panova
- Belozersky Institute of Physico-Chemical Biology, Department of Materials Science, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.G.F.); (M.V.K.); (E.A.P.)
| | - Elena V. Shmendel
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Mikhail A. Maslov
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, 119571 Moscow, Russia; (P.A.P.); (D.M.M.); (E.V.S.); (M.A.M.)
| | - Sergey E. Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Department of Materials Science, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; (A.G.F.); (M.V.K.); (E.A.P.)
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Grigory A. Stepanov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| | - Oleg V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (D.N.A.); (A.S.D.); (A.M.M.); (G.A.S.)
| |
Collapse
|
19
|
Kenison JE, Stevens NA, Quintana FJ. Therapeutic induction of antigen-specific immune tolerance. Nat Rev Immunol 2024; 24:338-357. [PMID: 38086932 PMCID: PMC11145724 DOI: 10.1038/s41577-023-00970-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 05/04/2024]
Abstract
The development of therapeutic approaches for the induction of robust, long-lasting and antigen-specific immune tolerance remains an important unmet clinical need for the management of autoimmunity, allergy, organ transplantation and gene therapy. Recent breakthroughs in our understanding of immune tolerance mechanisms have opened new research avenues and therapeutic opportunities in this area. Here, we review mechanisms of immune tolerance and novel methods for its therapeutic induction.
Collapse
Affiliation(s)
- Jessica E Kenison
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nikolas A Stevens
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
20
|
Nele V, Campani V, Alia Moosavian S, De Rosa G. Lipid nanoparticles for RNA delivery: Self-assembling vs driven-assembling strategies. Adv Drug Deliv Rev 2024; 208:115291. [PMID: 38514018 DOI: 10.1016/j.addr.2024.115291] [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: 12/22/2023] [Revised: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
Among non-viral vectors, lipid nanovectors are considered the gold standard for the delivery of RNA therapeutics. The success of lipid nanoparticles for RNA delivery, with three products approved for human use, has stimulated further investigation into RNA therapeutics for different pathologies. This requires decoding the pathological intracellular processes and tailoring the delivery system to the target tissue and cells. The complexity of the lipid nanovectors morphology originates from the assembling of the lipidic components, which can be elicited by various methods able to drive the formation of nanoparticles with the desired organization. In other cases, pre-formed nanoparticles can be mixed with RNA to induce self-assembly and structural reorganization into RNA-loaded nanoparticles. In this review, the most relevant lipid nanovectors and their potentialities for RNA delivery are described on the basis of the assembling mechanism and of the particle architecture.
Collapse
Affiliation(s)
- Valeria Nele
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Virginia Campani
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Seyedeh Alia Moosavian
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano, 49 80131 Naples, Italy.
| |
Collapse
|
21
|
Zhang X, Ma Y, Shi Y, Jiang L, Wang L, Ur Rashid H, Yuan M, Liu X. Advances in liposomes loaded with photoresponse materials for cancer therapy. Biomed Pharmacother 2024; 174:116586. [PMID: 38626516 DOI: 10.1016/j.biopha.2024.116586] [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: 02/22/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/18/2024] Open
Abstract
Cancer treatment is presently a significant challenge in the medical domain, wherein the primary modalities of intervention include chemotherapy, radiation therapy and surgery. However, these therapeutic modalities carry side effects. Photothermal therapy (PTT) and photodynamic therapy (PDT) have emerged as promising modalities for the treatment of tumors in recent years. Phototherapy is a therapeutic approach that involves the exposure of materials to specific wavelengths of light, which can subsequently be converted into either heat or Reactive Oxygen Species (ROS) to effectively eradicate cancer cells. Due to the hydrophobicity and lack of targeting of many photoresponsive materials, the use of nano-carriers for their transportation has been extensively explored. Among these nanocarriers, liposomes have been identified as an effective drug delivery system due to their controllability and availability in the biomedical field. By binding photoresponsive materials to liposomes, it is possible to reduce the cytotoxicity of the material and regulate drug release and accumulation at the tumor site. This article provides a comprehensive review of the progress made in cancer therapy using photoresponsive materials loaded onto liposomes. Additionally, the article discusses the potential synergistic treatment through the combination of phototherapy with chemo/immuno/gene therapy using liposomes.
Collapse
Affiliation(s)
- Xianwei Zhang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Youfu Ma
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Yenong Shi
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Lihe Jiang
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Lisheng Wang
- Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Pelotas, RS 96010-900, Brazil
| | - Haroon Ur Rashid
- Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Pelotas, RS 96010-900, Brazil
| | - Mingqing Yuan
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| | - Xu Liu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| |
Collapse
|
22
|
Homma K, Miura Y, Kobayashi M, Chintrakulchai W, Toyoda M, Ogi K, Michinishi J, Ohtake T, Honda Y, Nomoto T, Takemoto H, Nishiyama N. Fine tuning of the net charge alternation of polyzwitterion surfaced lipid nanoparticles to enhance cellular uptake and membrane fusion potential. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2338785. [PMID: 38646148 PMCID: PMC11028023 DOI: 10.1080/14686996.2024.2338785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/25/2024] [Indexed: 04/23/2024]
Abstract
Lipid nanoparticles (LNPs) coated with functional and biocompatible polymers have been widely used as carriers to deliver oligonucleotide and messenger RNA therapeutics to treat diseases. Poly(ethylene glycol) (PEG) is a representative material used for the surface coating, but the PEG surface-coated LNPs often have reduced cellular uptake efficiency and pharmacological activity. Here, we demonstrate the effect of pH-responsive ethylenediamine-based polycarboxybetaines with different molecular weights as an alternative structural component to PEG for the coating of LNPs. We found that appropriate tuning of the molecular weight around polycarboxybetaine-modified LNP, which incorporated small interfering RNA, could enhance the cellular uptake and membrane fusion potential in cancerous pH condition, thereby facilitating the gene silencing effect. This study demonstrates the importance of the design and molecular length of polymers on the LNP surface to provide effective drug delivery to cancer cells.
Collapse
Affiliation(s)
- Keitaro Homma
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Motoaki Kobayashi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Wanphiwat Chintrakulchai
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Masahiro Toyoda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Koichi Ogi
- I&S Department, Corporate R&D division, NOF CORPORATION, Kanagawa, Japan
| | - Junya Michinishi
- I&S Department, Corporate R&D division, NOF CORPORATION, Kanagawa, Japan
| | - Tomoyuki Ohtake
- I&S Department, Corporate R&D division, NOF CORPORATION, Kanagawa, Japan
| | - Yuto Honda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kanagawa, Japan
| | - Takahiro Nomoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyasu Takemoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Kanagawa, Japan
- Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kanagawa, Japan
| |
Collapse
|
23
|
Chen J, Wang B, Wang Y, Radermacher H, Qi J, Momoh J, Lammers T, Shi Y, Rix A, Kiessling F. mRNA Sonotransfection of Tumors with Polymeric Microbubbles: Co-Formulation versus Co-Administration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306139. [PMID: 38342634 PMCID: PMC11022722 DOI: 10.1002/advs.202306139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/24/2024] [Indexed: 02/13/2024]
Abstract
Despite its high potential, non-viral gene therapy of cancer remains challenging due to inefficient nucleic acid delivery. Ultrasound (US) with microbubbles (MB) can open biological barriers and thus improve DNA and mRNA passage. Polymeric MB are an interesting alternative to clinically used lipid-coated MB because of their high stability, narrow size distribution, and easy functionalization. However, besides choosing the ideal MB, it remains unclear whether nanocarrier-encapsulated mRNA should be administered separately (co-administration) or conjugated to MB (co-formulation). Therefore, the impact of poly(n-butyl cyanoacrylate) MB co-administration with mRNA-DOTAP/DOPE lipoplexes or their co-formulation on the transfection of cancer cells in vitro and in vivo is analyzed. Sonotransfection improved mRNA delivery into 4T1 breast cancer cells in vitro with co-administration being more efficient than co-formulation. In vivo, the co-administration sonotransfection approach also resulted in higher transfection efficiency and reached deeper into the tumor tissue. On the contrary, co-formulation mainly promoted transfection of endothelial and perivascular cells. Furthermore, the co-formulation approach is much more dependent on the US trigger, resulting in significantly lower off-site transfection. Thus, the findings indicate that the choice of co-administration or co-formulation in sonotransfection should depend on the targeted cell population, tolerable off-site transfection, and the therapeutic purpose.
Collapse
Affiliation(s)
- Junlin Chen
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Bi Wang
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Yuchen Wang
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Harald Radermacher
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Jinwei Qi
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Jeffrey Momoh
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Twan Lammers
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Yang Shi
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Anne Rix
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular ImagingHelmholtz Institute for Biomedical EngineeringRWTH Aachen University52074AachenGermany
| |
Collapse
|
24
|
Wu H, Qin J, Ji W, Palupi NW, Yang M. Interaction between curcumin and ultrafiltered casein micelles or whey protein, and characteristics of their complexes. J Food Sci 2024; 89:1582-1598. [PMID: 38317423 DOI: 10.1111/1750-3841.16959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/23/2023] [Accepted: 01/11/2024] [Indexed: 02/07/2024]
Abstract
This work evaluated the interaction between micellar casein (MC) or whey protein (WP) in ultrafiltration retentate with curcumin (Cur), as well as the physicochemical and functional properties of Cur-MC and Cur-WP complexes. The MC had a higher affinity for Cur than WP, shown by higher binding constants of Cur-MC at various temperatures. Thermodynamic analysis of the binding process indicated that the interaction between Cur and MC or WP was hydrophobic in nature. Cur promoted the size and polydispersity index of MC and WP at 4 mM but did not alter the morphology of spray-dried MC and WP. The Cur-MC complexes showed better aqueous solubility at pH 2-3 and 6-10 compared to free MC. Combination with MC or WP improved the 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonate) radical scavenging activity of Cur. In addition, combination with MC and WP promoted cumulative release of Cur during simulated gastrointestinal digestion, especially for WP. Thus, MC and WP in retentates can be good alternative protein-based carriers for Cur delivery, whereas their complexes in powder form have good functional properties that could be used as active food ingredients in several food formulations. PRACTICAL APPLICATION: Microfiltration is a cheap and convenient approach that can be used to easily produce micellar casein (MC), with whey protein (WP) as one byproduct. In this study, we proved that MC and WP in retentates have strong interaction with curcumin (Cur), whereas their complexes have good functional properties. Thus, spray-dried MC-Cur or WP-Cur complexes could be used as active food ingredients in several food formulations.
Collapse
Affiliation(s)
- Hao Wu
- College of Science, Gansu Agricultural University, Lanzhou, China
| | - Juanjuan Qin
- College of Science, Gansu Agricultural University, Lanzhou, China
| | - Wei Ji
- College of Science, Gansu Agricultural University, Lanzhou, China
| | - Niken Widya Palupi
- Faculty of Agricultural Technology, University of Jember, Jember, Indonesia
| | - Min Yang
- College of Science, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
25
|
Slezak A, Chang K, Hossainy S, Mansurov A, Rowan SJ, Hubbell JA, Guler MO. Therapeutic synthetic and natural materials for immunoengineering. Chem Soc Rev 2024; 53:1789-1822. [PMID: 38170619 PMCID: PMC11557218 DOI: 10.1039/d3cs00805c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Immunoengineering is a rapidly evolving field that has been driving innovations in manipulating immune system for new treatment tools and methods. The need for materials for immunoengineering applications has gained significant attention in recent years due to the growing demand for effective therapies that can target and regulate the immune system. Biologics and biomaterials are emerging as promising tools for controlling immune responses, and a wide variety of materials, including proteins, polymers, nanoparticles, and hydrogels, are being developed for this purpose. In this review article, we explore the different types of materials used in immunoengineering applications, their properties and design principles, and highlight the latest therapeutic materials advancements. Recent works in adjuvants, vaccines, immune tolerance, immunotherapy, and tissue models for immunoengineering studies are discussed.
Collapse
Affiliation(s)
- Anna Slezak
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Kevin Chang
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Samir Hossainy
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Aslan Mansurov
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Stuart J Rowan
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Jeffrey A Hubbell
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| | - Mustafa O Guler
- The Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
26
|
Malakouti-Nejad M, Monti D, Burgalassi S, Bardania H, Elahi E, Morshedi D. A comparison between the effects of two liposome-encapsulated bevacizumab formulations on ocular neovascularization inhibition. Colloids Surf B Biointerfaces 2024; 234:113708. [PMID: 38141384 DOI: 10.1016/j.colsurfb.2023.113708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/26/2023] [Accepted: 12/12/2023] [Indexed: 12/25/2023]
Abstract
Bevacizumab (BVZ), an anti-VEGF antibody, has demonstrated reliable outcomes in the treatment of irritating ocular neovascularization. Frequent intravitreal injections are necessitated due to rapid clearance and short local accessibility. We recruited liposome as a highly prevailing drug delivery system to enhance drug availability. Two liposome formulations were characterized and their in vitro stability was analyzed. The toxicity of the formulations on some ocular cell lines was also evaluated. In addition, the anti-angiogenic effects of formulations were examined. Drug permeation was measured across ARPE-19 and HCE cell lines as in vitro cellular barrier models. Results revealed that NLP-DOPE-BVZ acquired high stability at 4 °C, 24 °C, and 37 °C for 45 days. It also showed more capacity to entrap BVZ in NLP-DOPE-BVZ (DEE% 69.1 ± 1.4 and DLE% 55.66 ± 1.15) as compared to NLP-BVZ (DEE% 43.57 ± 14.64, and DLE% 37.72 ± 12.01). Although both formulations inhibited the migration and proliferation of HUVECs, NLP-DOPE-BVZ was more effective at inhibiting angiogenesis. Furthermore, NLP-DOPE-BVZ better crossed our established barrier cellular models. Based on the findings, the inclusion of DOPE in NLPs has significantly enhanced the features of drug carriers. This makes them a potential candidate for treating ocular neovascularization and other related ailments.
Collapse
Affiliation(s)
- Maryam Malakouti-Nejad
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Daniela Monti
- Department of Pharmacy, University of Pisa, via Bonanno 33, 56126 Pisa, Italy
| | - Susi Burgalassi
- Department of Pharmacy, University of Pisa, via Bonanno 33, 56126 Pisa, Italy
| | - Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Dina Morshedi
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| |
Collapse
|
27
|
Delehedde C, Ciganek I, Laroui N, Rameix N, Perche F, Pichon C. Messenger RNA Lipid-Based Nanoparticles: Optimization of Formulations in the Lab. Methods Mol Biol 2024; 2786:255-287. [PMID: 38814399 DOI: 10.1007/978-1-0716-3770-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Among the large variety of messenger RNA (mRNA) delivery systems, those developed with lipid-based formulations were the most widely used and efficient. In our lab, we produced different mRNA formulations made with liposomes, hybrid lipid polymer, and lipid nanoparticles. Our formulations were made with lipids bearing imidazole groups that trigger the endosomal escape of nanoparticles once protonated inside the mild acidic milieu of endosomes upon their cell uptake. Herein, we describe protocols that we used to produce, optimize, and characterize those formulations. The transfection efficiency is influenced by various factors including the physicochemical parameters of the nanoparticles, their efficiency to be internalized in cells, and their intracellular routing as well as their capacity to induce immune system sensors. We provide details on how to quantify the amount of mRNA nanoparticles uptake by cells and evaluate the acidity of the intracellular compartments where they are located, to investigate the endosomal escape, and to assess the activation of innate immune sensors as phosphorylation of PKR hampering mRNA translation.
Collapse
Affiliation(s)
- Christophe Delehedde
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Ivan Ciganek
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Inserm, ART-ARNm Inserm US55, Orléans, France
| | - Nabila Laroui
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
- Inserm, ART-ARNm Inserm US55, Orléans, France
| | - Nathalie Rameix
- Sanofi R&D, Integrated Drug Discovery, Chilly-Mazarin, France
| | - Federico Perche
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France
| | - Chantal Pichon
- Innovative Therapies & Nanomedicine, Centre de Biophysique Moléculaire CNRS UPR01, Orléans, France.
- Orléans University, Château de la Source, Orleans, France.
- Institut Universitaire de France, Paris, France.
- Inserm, ART-ARNm Inserm US55, Orléans, France.
| |
Collapse
|
28
|
Gomes AA, Valverde TM, Machado VDO, do Nascimento da Silva E, Fagundes DA, Oliveira FDP, Freitas ETF, Ardisson JD, Ferreira JMDF, Oliveira JADC, Gomes ER, Rodrigues CF, de Goes AM, Domingues RZ, Andrade ÂL. Heating Capacity and Biocompatibility of Hybrid Nanoparticles for Magnetic Hyperthermia Treatment. Int J Mol Sci 2023; 25:493. [PMID: 38203662 PMCID: PMC10779024 DOI: 10.3390/ijms25010493] [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: 10/10/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 01/12/2024] Open
Abstract
Cancer is one of the deadliest diseases worldwide and has been responsible for millions of deaths. However, developing a satisfactory smart multifunctional material combining different strategies to kill cancer cells poses a challenge. This work aims at filling this gap by developing a composite material for cancer treatment through hyperthermia and drug release. With this purpose, magnetic nanoparticles were coated with a polymer matrix consisting of poly (L-co-D,L lactic acid-co-trimethylene carbonate) and a poly(ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) triblock copolymer. High-resolution transmission electron microscopy and selected area electron diffraction confirmed magnetite to be the only iron oxide in the sample. Cytotoxicity and heat release assays on the hybrid nanoparticles were performed here for the first time. The heat induction results indicate that these new magnetic hybrid nanoparticles are capable of increasing the temperature by more than 5 °C, the minimal temperature rise required for being effectively used in hyperthermia treatments. The biocompatibility assays conducted under different concentrations, in the presence and in the absence of an external alternating current magnetic field, did not reveal any cytotoxicity. Therefore, the overall results indicate that the investigated hybrid nanoparticles have a great potential to be used as carrier systems for cancer treatment by hyperthermia.
Collapse
Affiliation(s)
- Aline Alexandrina Gomes
- Departamento de Química, Instituto de Ciências Exatas e Biológicas (ICEB), Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (A.A.G.); (V.d.O.M.); (E.d.N.d.S.)
| | - Thalita Marcolan Valverde
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (T.M.V.); (C.F.R.)
| | - Vagner de Oliveira Machado
- Departamento de Química, Instituto de Ciências Exatas e Biológicas (ICEB), Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (A.A.G.); (V.d.O.M.); (E.d.N.d.S.)
| | - Emanueli do Nascimento da Silva
- Departamento de Química, Instituto de Ciências Exatas e Biológicas (ICEB), Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (A.A.G.); (V.d.O.M.); (E.d.N.d.S.)
| | - Daniele Alves Fagundes
- Laboratório de Física Aplicada, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN/CNEN), Belo Horizonte 31270-901, MG, Brazil; (D.A.F.); (F.d.P.O.); (J.D.A.)
| | - Fernanda de Paula Oliveira
- Laboratório de Física Aplicada, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN/CNEN), Belo Horizonte 31270-901, MG, Brazil; (D.A.F.); (F.d.P.O.); (J.D.A.)
| | | | - José Domingos Ardisson
- Laboratório de Física Aplicada, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN/CNEN), Belo Horizonte 31270-901, MG, Brazil; (D.A.F.); (F.d.P.O.); (J.D.A.)
| | - José Maria da Fonte Ferreira
- Departamento de Engenharia de Materiais e Cerâmica (CICECO), Universidade de Aveiro (UA), 3810193 Aveiro, Portugal;
| | - Junnia Alvarenga de Carvalho Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| | - Eliza Rocha Gomes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| | - Caio Fabrini Rodrigues
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil; (T.M.V.); (C.F.R.)
| | - Alfredo Miranda de Goes
- Departamento de Patologia Geral, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| | - Rosana Zacarias Domingues
- Departamento de Química, Instituto de Ciências Exatas (ICEx), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| | - Ângela Leão Andrade
- Departamento de Química, Instituto de Ciências Exatas e Biológicas (ICEB), Universidade Federal de Ouro Preto (UFOP), Ouro Preto 35400-000, MG, Brazil; (A.A.G.); (V.d.O.M.); (E.d.N.d.S.)
| |
Collapse
|
29
|
Scheeder A, Brockhoff M, Ward EN, Kaminski Schierle GS, Mela I, Kaminski CF. Molecular Mechanisms of Cationic Fusogenic Liposome Interactions with Bacterial Envelopes. J Am Chem Soc 2023; 145:28240-28250. [PMID: 38085801 PMCID: PMC10755748 DOI: 10.1021/jacs.3c11463] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/28/2023]
Abstract
Although fusogenic liposomes offer a promising approach for the delivery of antibiotic payloads across the cell envelope of Gram-negative bacteria, there is still a limited understanding of the individual nanocarrier interactions with the bacterial target. Using super-resolution microscopy, we characterize the interaction dynamics of positively charged fusogenic liposomes with Gram-negative (Escherichia coli) and Gram-positive (Bacillus subtilis) bacteria. The liposomes merge with the outer membrane (OM) of Gram-negative bacteria, while attachment or lipid internalization is observed in Gram-positive cells. Employing total internal reflection fluorescence microscopy, we demonstrated liposome fusion with model supported lipid bilayers. For whole E. coli cells, however, we observed heterogeneous membrane integrations, primarily involving liposome attachment and hemifusion events. With increasing lipopolysaccharide length, the likelihood of full-fusion events was reduced. The integration of artificial lipids into the OM of Gram-negative cells led to membrane destabilization, resulting in decreased bacterial vitality, membrane detachment, and improved codelivery of vancomycin─an effective antibiotic against Gram-positive cells. These findings provide significant insights into the interactions of individual nanocarriers with bacterial envelopes at the single-cell level, uncovering effects that would be missed in bulk measurements. This highlights the importance of conducting single-particle and single-cell investigations to assess the performance of next-generation drug delivery platforms.
Collapse
Affiliation(s)
- Anna Scheeder
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Marius Brockhoff
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Edward N. Ward
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Gabriele S. Kaminski Schierle
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| | - Ioanna Mela
- Department
of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K.
| | - Clemens F. Kaminski
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.
| |
Collapse
|
30
|
Walker AJ, Graham C, Greenwood M, Woodall M, Maeshima R, O’Hara-Wright M, Sanz DJ, Guerrini I, Aldossary AM, O’Callaghan C, Baines DL, Harrison PT, Hart SL. Molecular and functional correction of a deep intronic splicing mutation in CFTR by CRISPR-Cas9 gene editing. Mol Ther Methods Clin Dev 2023; 31:101140. [PMID: 38027060 PMCID: PMC10661860 DOI: 10.1016/j.omtm.2023.101140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/17/2023] [Indexed: 12/01/2023]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the CFTR gene. The 10th most common mutation, c.3178-2477C>T (3849+10kb C>T), involves a cryptic, intronic splice site. This mutation was corrected in CF primary cells homozygous for this mutation by delivering pairs of guide RNAs (gRNAs) with Cas9 protein in ribonucleoprotein (RNP) complexes that introduce double-strand breaks to flanking sites to excise the 3849+10kb C>T mutation, followed by DNA repair by the non-homologous end-joining pathway, which functions in all cells of the airway epithelium. RNP complexes were delivered to CF basal epithelial cell by a non-viral, receptor-targeted nanocomplex comprising a formulation of targeting peptides and lipids. Canonical CFTR mRNA splicing was, thus, restored leading to the restoration of CFTR protein expression with concomitant restoration of electrophysiological function in airway epithelial air-liquid interface cultures. Off-target editing was not detected by Sanger sequencing of in silico-selected genomic sites with the highest sequence similarities to the gRNAs, although more sensitive unbiased whole genome sequencing methods would be required for possible translational developments. This approach could potentially be used to correct aberrant splicing signals in several other CF mutations and other genetic disorders where deep-intronic mutations are pathogenic.
Collapse
Affiliation(s)
- Amy J. Walker
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Carina Graham
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Miriam Greenwood
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Maximillian Woodall
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
| | - Ruhina Maeshima
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Michelle O’Hara-Wright
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - David J. Sanz
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Ileana Guerrini
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Ahmad M. Aldossary
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Christopher O’Callaghan
- Infection, Immunity & Inflammation Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Deborah L. Baines
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
| | - Patrick T. Harrison
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Stephen L. Hart
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
31
|
Bolsoni J, Liu D, Mohabatpour F, Ebner R, Sadhnani G, Tafech B, Leung J, Shanta S, An K, Morin T, Chen Y, Arguello A, Choate K, Jan E, Ross CJ, Brambilla D, Witzigmann D, Kulkarni J, Cullis PR, Hedtrich S. Lipid Nanoparticle-Mediated Hit-and-Run Approaches Yield Efficient and Safe In Situ Gene Editing in Human Skin. ACS NANO 2023; 17:22046-22059. [PMID: 37918441 PMCID: PMC10655174 DOI: 10.1021/acsnano.3c08644] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
Despite exciting advances in gene editing, the efficient delivery of genetic tools to extrahepatic tissues remains challenging. This holds particularly true for the skin, which poses a highly restrictive delivery barrier. In this study, we ran a head-to-head comparison between Cas9 mRNA or ribonucleoprotein (RNP)-loaded lipid nanoparticles (LNPs) to deliver gene editing tools into epidermal layers of human skin, aiming for in situ gene editing. We observed distinct LNP composition and cell-specific effects such as an extended presence of RNP in slow-cycling epithelial cells for up to 72 h. While obtaining similar gene editing rates using Cas9 RNP and mRNA with MC3-based LNPs (10-16%), mRNA-loaded LNPs proved to be more cytotoxic. Interestingly, ionizable lipids with a pKa ∼ 7.1 yielded superior gene editing rates (55%-72%) in two-dimensional (2D) epithelial cells while no single guide RNA-dependent off-target effects were detectable. Unexpectedly, these high 2D editing efficacies did not translate to actual skin tissue where overall gene editing rates between 5%-12% were achieved after a single application and irrespective of the LNP composition. Finally, we successfully base-corrected a disease-causing mutation with an efficacy of ∼5% in autosomal recessive congenital ichthyosis patient cells, showcasing the potential of this strategy for the treatment of monogenic skin diseases. Taken together, this study demonstrates the feasibility of an in situ correction of disease-causing mutations in the skin that could provide effective treatment and potentially even a cure for rare, monogenic, and common skin diseases.
Collapse
Affiliation(s)
- Juliana Bolsoni
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Danny Liu
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Fatemeh Mohabatpour
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Ronja Ebner
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Gaurav Sadhnani
- Berlin
Institute of Health @ Charité Universitätsmedizin, Berlin 10117, Germany
| | - Belal Tafech
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Jerry Leung
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, BC, Canada
| | - Selina Shanta
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Kevin An
- NanoVation
Therapeutics, 2405 Wesbrook
Mall, Vancouver V6T 1Z3, BC, Canada
| | - Tessa Morin
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Yihang Chen
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, BC, Canada
| | - Alfonso Arguello
- University
of Montréal, Faculty of Pharmacy, Montréal H3T 1J4, Quebec, Canada
| | - Keith Choate
- Departments
of Dermatology, Genetics, and Pathology, Yale University School of Medicine, New Haven 06510, Connecticut, United States
| | - Eric Jan
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, BC, Canada
| | - Colin J.D. Ross
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Davide Brambilla
- University
of Montréal, Faculty of Pharmacy, Montréal H3T 1J4, Quebec, Canada
| | - Dominik Witzigmann
- NanoVation
Therapeutics, 2405 Wesbrook
Mall, Vancouver V6T 1Z3, BC, Canada
| | - Jayesh Kulkarni
- NanoVation
Therapeutics, 2405 Wesbrook
Mall, Vancouver V6T 1Z3, BC, Canada
| | - Pieter R. Cullis
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, BC, Canada
| | - Sarah Hedtrich
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
- Berlin
Institute of Health @ Charité Universitätsmedizin, Berlin 10117, Germany
- Department
of Infectious Diseases and Respiratory Medicine, Charité -
Universitätsmedizin Berlin, corporate
member of Freie Universität Berlin and Humboldt Universität, Berlin 10117, Germany
- Max-Delbrück
Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 13125, Germany
| |
Collapse
|
32
|
Kimura S, Harashima H. On the mechanism of tissue-selective gene delivery by lipid nanoparticles. J Control Release 2023; 362:797-811. [PMID: 37004796 DOI: 10.1016/j.jconrel.2023.03.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/04/2023]
Abstract
The era of nucleic acid nanomedicine has arrived, as evidenced by Patisiran, a small interfering RNA (siRNA) encapsulated lipid nanoparticle (LNP), and mRNA-loaded LNPs used in COVID-19 vaccines. The diversity of nano-designs for delivering nucleic acid molecules tested in Phase II/III clinical trials reflects the potential of these technologies. These breakthroughs in non-viral gene delivery, including the use of LNPs, have attracted substantial interest worldwide for developing more effective drugs. A next step in this field is to target tissues other than the liver, which requires significant research efforts and material development. However, mechanistic studies in this area are lacking. This study compares two types of LNPs with different tissue-selectivity for delivering plasmid DNA (pDNA), one being liver-selective and the other spleen-selective, in an effort to understand the mechanisms responsible for differences in gene expression of delivered genes. We observed little difference in the biodistribution of these two LNPs despite the 100-1000-fold differences in gene expression. We then quantified the amount of delivered pDNA and mRNA expression in each tissue by quantitative real-time PCR (qPCR) to evaluate various intracellular processes, such as nuclear delivery, transcription and translation. The results showed a >100-fold difference in the translation step but there were little differences in amount of pDNA delivered to the nucleus or the amount of mRNA expression for the two LNP deliveries. Our findings suggest that endogenous factors affect gene expression efficiency not the extent of biodistribution.
Collapse
Affiliation(s)
- Seigo Kimura
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan; Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
33
|
Gandek TB, van der Koog L, Nagelkerke A. A Comparison of Cellular Uptake Mechanisms, Delivery Efficacy, and Intracellular Fate between Liposomes and Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2300319. [PMID: 37384827 PMCID: PMC11469107 DOI: 10.1002/adhm.202300319] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
A key aspect for successful drug delivery via lipid-based nanoparticles is their internalization in target cells. Two prominent examples of such drug delivery systems are artificial phospholipid-based carriers, such as liposomes, and their biological counterparts, the extracellular vesicles (EVs). Despite a wealth of literature, it remains unclear which mechanisms precisely orchestrate nanoparticle-mediated cargo delivery to recipient cells and the subsequent intracellular fate of therapeutic cargo. In this review, internalization mechanisms involved in the uptake of liposomes and EVs by recipient cells are evaluated, also exploring their intracellular fate after intracellular trafficking. Opportunities are highlighted to tweak these internalization mechanisms and intracellular fates to enhance the therapeutic efficacy of these drug delivery systems. Overall, literature to date shows that both liposomes and EVs are predominantly internalized through classical endocytosis mechanisms, sharing a common fate: accumulation inside lysosomes. Studies tackling the differences between liposomes and EVs, with respect to cellular uptake, intracellular delivery and therapy efficacy, remain scarce, despite its importance for the selection of an appropriate drug delivery system. In addition, further exploration of functionalization strategies of both liposomes and EVs represents an important avenue to pursue in order to control internalization and fate, thereby improving therapeutic efficacy.
Collapse
Affiliation(s)
- Timea B. Gandek
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| | - Luke van der Koog
- Molecular PharmacologyGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB10Groningen9700 ADThe Netherlands
| | - Anika Nagelkerke
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| |
Collapse
|
34
|
Fus-Kujawa A, Mendrek B, Bajdak-Rusinek K, Diak N, Strzelec K, Gutmajster E, Janelt K, Kowalczuk A, Trybus A, Rozwadowska P, Wojakowski W, Gawron K, Sieroń AL. Gene-repaired iPS cells as novel approach for patient with osteogenesis imperfecta. Front Bioeng Biotechnol 2023; 11:1205122. [PMID: 37456734 PMCID: PMC10348904 DOI: 10.3389/fbioe.2023.1205122] [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: 04/13/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction: The benefits of patient's specific cell/gene therapy have been reported in relation to numerous genetic related disorders including osteogenesis imperfecta (OI). In osteogenesis imperfecta particularly also a drug therapy based on the administration of bisphosphonates partially helped to ease the symptoms. Methods: In this controlled trial, fibroblasts derived from patient diagnosed with OI type II have been successfully reprogrammed into induced Pluripotent Stem cells (iPSCs) using Yamanaka factors. Those cells were subjected to repair mutations found in the COL1A1 gene using homologous recombination (HR) approach facilitated with star polymer (STAR) as a carrier of the genetic material. Results: Delivery of the correct linear DNA fragment to the osteogenesis imperfecta patient's cells resulted in the repair of the DNA mutation with an 84% success rate. IPSCs showed 87% viability after STAR treatment and 82% with its polyplex. Discussion: The use of novel polymer Poly[N,N-Dimethylaminoethyl Methacrylate-co-Hydroxyl-Bearing Oligo(Ethylene Glycol) Methacrylate] Arms (P(DMAEMA-co-OEGMA-OH) with star-like structure has been shown as an efficient tool for nucleic acids delivery into cells (Funded by National Science Centre, Contract No. UMO-2020/37/N/NZ2/01125).
Collapse
Affiliation(s)
- Agnieszka Fus-Kujawa
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Barbara Mendrek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Karolina Bajdak-Rusinek
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Natalia Diak
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Karolina Strzelec
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Ewa Gutmajster
- Biotechnology Centre, Silesian University of Technology, Gliwice, Poland
| | - Kamil Janelt
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agnieszka Kowalczuk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Anna Trybus
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
- Students Scientific Society, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Patrycja Rozwadowska
- Department of Medical Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
- Students Scientific Society, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Wojciech Wojakowski
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
| | - Katarzyna Gawron
- Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Aleksander L. Sieroń
- Formerly Department of Molecular Biology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
35
|
Montanari E, Krupke H, Leroux JC. Engineering Lipid Spherulites for the Sustained Release of Highly Dosed Small Hydrophilic Compounds. Adv Healthc Mater 2023; 12:e2202249. [PMID: 36571233 PMCID: PMC11469156 DOI: 10.1002/adhm.202202249] [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: 09/02/2022] [Revised: 12/21/2022] [Indexed: 12/27/2022]
Abstract
Currently, there is a lack of parenteral sustained release formulations for the delivery of highly dosed small hydrophilic drugs. Therefore, parenteral lipid spherulites are engineered capable of entrapping large amounts of such compounds and spontaneously releasing them in a sustained fashion. A library of spherulites is prepared with a simple green process, using phosphatidylcholine (PC) and/or phosphatidylethanolamine (PE), nonionic surfactants and water. The vesicle formulations exhibiting appropriate size distribution and morphology are selected and loaded with 4,6-di-O-(methoxy-diethyleneglycol)-myo-inositol-1,2,3,5-tetrakis(phosphate), ((OEG2 )2 -IP4), an inositol phosphate derivative currently under clinical evaluation for the treatment of aortic valve stenosis. The loading efficiency of spherulites is up to 12.5-fold higher than that of liposomes produced with the same materials. While the PC-containing vesicles showed high stability, the PE spherulites gradually lost their multilayer organization upon dilution, triggering the active pharmaceutical ingredient (API) release over time. In vitro experiments and pharmacokinetic studies in rats demonstrated the ability of PE spherulites to increase the systemic exposure of (OEG2 )2 -IP4 up to 3.1-fold after subcutaneous injection, and to completely release their payload within 3-4 d. In conclusion, PE spherulites represent a promising lipid platform for the extravascular parenteral administration of highly dosed small hydrophilic drugs.
Collapse
Affiliation(s)
- Elita Montanari
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, 8093, Switzerland
| | - Hanna Krupke
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, 8093, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, 8093, Switzerland
| |
Collapse
|
36
|
Ghanem R, Berchel M, Haute T, Buin X, Laurent V, Youf R, Bouraoui A, Le Gall T, Jaffrès PA, Montier T. Gene transfection using branched cationic amphiphilic compounds for an aerosol administration in cystic fibrosis context. Int J Pharm 2023; 631:122491. [PMID: 36529361 DOI: 10.1016/j.ijpharm.2022.122491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/29/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
For cystic fibrosis gene therapy, the aerosolization of genetic materials is the most relevant delivery strategy to reach the airway epithelium. However, aerosolized formulations have to resist shear forces while maintaining the integrity of plasmid DNA (pDNA) during its journey from the nebulization to the epithelial cells. Herein, we compared the efficiency of gene delivery by aerosolization of two types of formulations: (i) BSV163, a branched cationic amphiphilic compound, co-formulated with different DOPE ratios (mol/mol) and DMPE-PEG5000 and (ii) 25 KDa branched polyethylenimine (b-PEI)-based formulation used as control. This study also aims to determine whether BSV163-based formulations possess the ability to resist the nebulization mechanisms and protect the nucleic acids (pDNA) cargo. Therefore, two CpG free plasmids (pGM144 or pGM169) encoding either the luciferase reporter gene or hCFTR respectively were used. Air-Liquid Interface (ALI) cell-culture was selected as an in-vitro model for aerosol experiments due to its closer analogy with in vivo morphology. Results highlighted that DOPE ratio influences the capacity of the BSV163 based-formulations to mediate high transfection efficacies. Furthermore, we proved that addition of DMPE-PEG5000 upon the formation of the BSV163/DOPE (1/1) lipid film instead of post-insertion led to a higher transgene expression. The aerosolization of this formulation on ALI cell-culture was more efficient than the use of b-PEI-based formulation.
Collapse
Affiliation(s)
- Rosy Ghanem
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200 Brest, France
| | - Mathieu Berchel
- Univ Brest, CNRS, CEMCA UMR 65216, Avenue Victor, Le Gorgeu, F-29238 Brest, France
| | - Tanguy Haute
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200 Brest, France
| | - Xavier Buin
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200 Brest, France
| | | | - Raphaëlle Youf
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200 Brest, France
| | - Amal Bouraoui
- Univ Brest, CNRS, CEMCA UMR 65216, Avenue Victor, Le Gorgeu, F-29238 Brest, France
| | - Tony Le Gall
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200 Brest, France
| | - Paul-Alain Jaffrès
- Univ Brest, CNRS, CEMCA UMR 65216, Avenue Victor, Le Gorgeu, F-29238 Brest, France
| | - Tristan Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200 Brest, France; CHRU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200 Brest, France.
| |
Collapse
|
37
|
One-Step Pharmaceutical Preparation of PEG-Modified Exosomes Encapsulating Anti-Cancer Drugs by a High-Pressure Homogenization Technique. Pharmaceuticals (Basel) 2023; 16:ph16010108. [PMID: 36678605 PMCID: PMC9865360 DOI: 10.3390/ph16010108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/04/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
The use of exosomes encapsulating therapeutic agents for the treatment of diseases is of increasing interest. However, some concerns such as limited efficiency and scalability of conventional drug encapsulation methods to exosomes have still remained; thus, a new approach that enables encapsulation of therapeutic agents with superior efficiency and scalability is required. Herein, we used RAW264 macrophage cell-derived exosomes (RAW-Exos) and demonstrated that high-pressure homogenization (HPH) using a microfluidizer decreased their particle size without changing their morphology, the amount of exosomal marker proteins, and cellular uptake efficiency into RAW264 and colon-26 cancer cells. Moreover, HPH allowed for modification of polyethylene glycol (PEG)-conjugated lipids onto RAW-Exos, as well as encapsulation of the anti-cancer agent doxorubicin. Importantly, the doxorubicin encapsulation efficiency became higher upon increasing the process pressure and simultaneous HPH with PEG-lipids. Moreover, treatment with PEG-modified RAW-Exos encapsulating doxorubicin significantly suppressed tumor growth in colon-26-bearing mice. Taken together, these results suggest that HPH using a microfluidizer could be useful to prepare PEG-modified Exos encapsulating anti-cancer drugs via a one-step pharmaceutical process, and that the prepared functional Exos could be applied for the treatment of cancer in vivo.
Collapse
|
38
|
Andretto V, Repellin M, Pujol M, Almouazen E, Sidi-Boumedine J, Granjon T, Zhang H, Remaut K, Jordheim LP, Briançon S, Keil IS, Vascotto F, Walzer KC, Sahin U, Haas H, Kryza D, Lollo G. Hybrid core-shell particles for mRNA systemic delivery. J Control Release 2023; 353:1037-1049. [PMID: 36442614 DOI: 10.1016/j.jconrel.2022.11.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/15/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
mRNA based infectious disease vaccines have opened the venue for development of novel nucleic acids-based therapeutics. For all mRNA therapeutics dedicated delivery systems are required, where different functionalities and targeting abilities need to be optimized for the respective applications. One option for advanced formulations with tailored properties are lipid-polymer hybrid nanoparticles with complex nanostructure, which allow to combine features of several already well described nucleic acid delivery systems. Here, we explored hyaluronic acid (HA) as coating of liposome-mRNA complexes (LRCs) to investigate effects of the coating on surface charge, physicochemical characteristics and biological activity. HA was electrostatically attached to positively charged complexes, forming hybrid LRCs (HLRCs). At different N/P ratios, physico-chemical characterization of the two sets of particles showed similarity in size (around 200 nm) and mRNA binding abilities, while the presence of the HA shell conferred a negative surface charge to otherwise positive complexes. High transfection efficiency of LRCs and HLRCs in vitro has been obtained in THP-1 and human monocytes derived from PBMC, an interesting target cell population for cancer and immune related pathologies. In mice, quantitative biodistribution of radiolabeled LRC and HLRC particles, coupled with bioluminescence studies to detect the protein translation sites, hinted towards both particles' accumulation in the hepatic reticuloendothelial system (RES). mRNA translated proteins though was found mainly in the spleen, a major source for immune cells, with preference for expression in macrophages. The results showed that surface modifications of liposome-mRNA complexes can be used to fine-tune nanoparticle physico-chemical characteristics. This provides a tool for assembly of stable and optimized nanoparticles, which are prerequisite for future therapeutic interventions using mRNA-based nanomedicines.
Collapse
Affiliation(s)
- Valentina Andretto
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France
| | - Mathieu Repellin
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France
| | - Marine Pujol
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France
| | - Eyad Almouazen
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France
| | - Jacqueline Sidi-Boumedine
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France
| | - Thierry Granjon
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires, ICBMS UMR 5246, Université de Lyon, Université Lyon 1, CNRS, F-69622 Lyon, France
| | - Heyang Zhang
- Ghent Research Group on Nanomedicine, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicine, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lars Petter Jordheim
- Univ. Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon 69008, France
| | - Stéphanie Briançon
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France
| | - Isabell Sofia Keil
- TRON Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Fulvia Vascotto
- TRON Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Mainz, Germany
| | | | - Ugur Sahin
- BioNTech SE, An der Goldgrube 12, 55131 Mainz, Germany
| | - Heinrich Haas
- BioNTech SE, An der Goldgrube 12, 55131 Mainz, Germany
| | - David Kryza
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France; Hospices Civils de Lyon, 69437 Lyon, France
| | - Giovanna Lollo
- Univ. Lyon, Université Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007, 43 Boulevard du 11 Novembre 1918, F-69622, Villeurbanne, France.
| |
Collapse
|
39
|
Licensed liposomal vaccines and adjuvants in the antigen delivery system. BIOTECHNOLOGIA 2022; 103:409-423. [PMID: 36685697 PMCID: PMC9837556 DOI: 10.5114/bta.2022.120709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/10/2022] [Accepted: 08/02/2022] [Indexed: 12/28/2022] Open
Abstract
Liposomes (LSs) are promising nanoparticles with unique properties such as controlled nanosize, large surface area, increased reactivity, and ability to undergo modification. Worldwide, licensed liposomal forms of antibiotics, hormones, antioxidants, cytostatics, ophthalmic drugs, etc., are available on the pharmaceutical market. This review focuses on the adjuvant properties of LSs in the production of vaccines (VACs). LS-VACs have the following advantages: antigens with low immunogenicity can become highly immunogenic; LSs can include both hydrophilic and hydrophobic antigens; LSs allow to achieve a prolonged specific action of antibodies; and LSs reduce the toxicity and pyrogenicity of encapsulated antigens and adjuvants. The immune response is influenced by the composition of the liposomal membrane, physicochemical characteristics of lipids, antigen localization in LSs, interaction of LSs with complement, and a number of proteins, which leads to opsonization. The major requirements for adjuvants are their ability to enhance the immune response, biodegradability, and elimination from the organism, and LSs fully meet these requirements. The effectiveness and safety of LSs as carriers in the antigen delivery system have been proven by the long-term clinical use of licensed vaccines against hepatitis A, influenza, herpes zoster, malaria, and COVID-19.
Collapse
|
40
|
Vysochinskaya V, Shishlyannikov S, Zabrodskaya Y, Shmendel E, Klotchenko S, Dobrovolskaya O, Gavrilova N, Makarova D, Plotnikova M, Elpaeva E, Gorshkov A, Moshkoff D, Maslov M, Vasin A. Influence of Lipid Composition of Cationic Liposomes 2X3-DOPE on mRNA Delivery into Eukaryotic Cells. Pharmaceutics 2022; 15:pharmaceutics15010008. [PMID: 36678637 PMCID: PMC9860636 DOI: 10.3390/pharmaceutics15010008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
The design of cationic liposomes for efficient mRNA delivery can significantly improve mRNA-based therapies. Lipoplexes based on polycationic lipid 1,26-bis(cholest-5-en-3β-yloxycarbonylamino)-7,11,16,20-tetraazahexacosane tetrahydrochloride (2X3) and helper lipid 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) were formulated in different molar ratios (1:1, 1:2, 1:3) to efficiently deliver model mRNAs to BHK-21 and A549. The objective of this study was to examine the effect of 2X3-DOPE composition as well as lipid-to-mRNA ratio (amino-to-phosphate group ratio, N/P) on mRNA transfection. We found that lipoplex-mediated transfection efficiency depends on both liposome composition and the N/P ratio. Lipoplexes with an N/P ratio of 10/1 showed nanometric hydrodynamic size, positive ζ potential, maximum loading, and transfection efficiency. Liposomes 2X3-DOPE (1:3) provided the superior delivery of both mRNA coding firefly luciferase and mRNA-eGFP into BHK-21 cells and A549 cells, compared with commercial Lipofectamine MessengerMax.
Collapse
Affiliation(s)
- Vera Vysochinskaya
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Ulitsa Polytechnicheskaya, 194064 St. Petersburg, Russia
- Correspondence:
| | - Sergey Shishlyannikov
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Ulitsa Polytechnicheskaya, 194064 St. Petersburg, Russia
| | - Yana Zabrodskaya
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Ulitsa Polytechnicheskaya, 194064 St. Petersburg, Russia
| | - Elena Shmendel
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 86 Vernadsky Ave, 119571 Moscow, Russia
| | - Sergey Klotchenko
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
| | - Olga Dobrovolskaya
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
| | - Nina Gavrilova
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
| | - Darya Makarova
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 86 Vernadsky Ave, 119571 Moscow, Russia
| | - Marina Plotnikova
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
| | - Ekaterina Elpaeva
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
| | - Andrey Gorshkov
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
| | - Dmitry Moshkoff
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Ulitsa Polytechnicheskaya, 194064 St. Petersburg, Russia
- Global Virus Network (GVN), 725 W Lombard St, Baltimore, MD 21201, USA
| | - Mikhail Maslov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, 86 Vernadsky Ave, 119571 Moscow, Russia
| | - Andrey Vasin
- Smorodintsev Research Institute of Influenza, 15/17 Ulitsa Professora Popova, 197376 St. Petersburg, Russia
- Institute of Biomedical Systems and Biotechnology, Peter the Great Saint Petersburg Polytechnic University, 29 Ulitsa Polytechnicheskaya, 194064 St. Petersburg, Russia
| |
Collapse
|
41
|
Nardo D, Pitts MG, Kaur R, Venditto VJ. In vivo assessment of triazine lipid nanoparticles as transfection agents for plasmid DNA. Biomater Sci 2022; 10:6968-6979. [PMID: 36222485 PMCID: PMC9729407 DOI: 10.1039/d2bm01289h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Non-viral vectors for in vivo delivery of plasmid DNA rely on optimized formulations to achieve robust transgene expression. Several cationic lipids have been developed to deliver nucleic acids, but most recent literature has focused on mRNA due to its increased expression profile and excluded plasmid DNA, which may have the advantage of being less immunogenic. In this study, we describe the in vivo evaluation of cationic triazine based lipids, previously prepared by our group. We identify one lipid with limited in vivo toxicity for studies to optimize the lipid formulations, which include an evaluation of the influence of PEG and helper lipids on transgene expression. We then demonstrate that lipoplexes, but not lipid nanoparticles, formed from triazine lipids achieve similar transgene expression levels as AAV vectors and offer enhanced expression as compared to a commercially available cationic lipid, DOTAP. Importantly, the lipid nanoparticles and lipoplexes induce minimal antibody profiles toward the expressed protein, while serving as a platform to induce robust antibody responses when directly delivering the protein. Collectively, these data demonstrate the potential for triazine based lipids as non-viral vectors for gene delivery, and highlights the need to optimize each formulation based on the exact contents to achieve enhanced transgene expression with plasmid DNA constructs.
Collapse
Affiliation(s)
- David Nardo
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, 40536, USA.
| | - Michelle G Pitts
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, 40536, USA.
| | - Rupinder Kaur
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, 40536, USA.
| | - Vincent J Venditto
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, 40536, USA.
| |
Collapse
|
42
|
Fachel FNS, Frâncio L, Poletto É, Schuh RS, Teixeira HF, Giugliani R, Baldo G, Matte U. Gene editing strategies to treat lysosomal disorders: The example of mucopolysaccharidoses. Adv Drug Deliv Rev 2022; 191:114616. [PMID: 36356930 DOI: 10.1016/j.addr.2022.114616] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 09/20/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
Lysosomal storage disorders are a group of progressive multisystemic hereditary diseases with a combined incidence of 1:4,800. Here we review the clinical and molecular characteristics of these diseases, with a special focus on Mucopolysaccharidoses, caused primarily by the lysosomal storage of glycosaminoglycans. Different gene editing techniques can be used to ameliorate their symptoms, using both viral and nonviral delivery methods. Whereas these are still being tested in animal models, early results of phase I/II clinical trials of gene therapy show how this technology may impact the future treatment of these diseases. Hurdles related to specific hard-to-reach organs, such as the central nervous system, heart, joints, and the eye must be tackled. Finally, the regulatory framework necessary to advance into clinical practice is also discussed.
Collapse
Affiliation(s)
- Flávia Nathiely Silveira Fachel
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, UFRGS, Porto Alegre, RS, Brazil
| | - Lariane Frâncio
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, UFRGS, Porto Alegre, RS, Brazil
| | - Édina Poletto
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Roselena Silvestri Schuh
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, UFRGS, Porto Alegre, RS, Brazil
| | - Helder Ferreira Teixeira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, UFRGS, Porto Alegre, RS, Brazil
| | - Roberto Giugliani
- Programa de Pós-Graduação em Genética e Biologia Molecular, UFRGS, Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Departamento de Genética, UFRGS, Porto Alegre, RS, Brazil
| | - Guilherme Baldo
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, UFRGS, Porto Alegre, RS, Brazil; Departamento de Fisiologia, UFRGS, Porto Alegre, RS, Brazil
| | - Ursula Matte
- Laboratório de Células, Tecidos e Genes - Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Genética e Biologia Molecular, UFRGS, Porto Alegre, RS, Brazil; Departamento de Genética, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
43
|
Mashima R, Takada S. Lipid Nanoparticles: A Novel Gene Delivery Technique for Clinical Application. Curr Issues Mol Biol 2022; 44:5013-5027. [PMID: 36286056 PMCID: PMC9600891 DOI: 10.3390/cimb44100341] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/08/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Lipid nanoparticles (LNPs) are an emerging vehicle for gene delivery that accommodate both nucleic acid and protein. Based on the experience of therapeutic liposomes, current LNPs have been developed based on the chemistry of lipids and RNA and on the biology of human disease. LNPs have been used for the development of Onpattro, an siRNA drug for transthyretin-mediated amyloidosis, in 2018. The subsequent outbreak of COVID-19 required a vaccine for its suppression. LNP-based vaccine production received much attention for this and resulted in great success. In this review, the essential technology of LNP gene delivery has been described according to the chemistry for LNP production and biology for its clinical application.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
- Correspondence:
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| |
Collapse
|
44
|
Development of Nanoemulsions for Wound Dressings Containing Cassia alata L. Leaf Extraction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4282678. [PMID: 36267084 PMCID: PMC9578847 DOI: 10.1155/2022/4282678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022]
Abstract
Natural polymer-based hydrogel films possess considerable potential for use in biomedical applications and are excellent for wound healing. The purpose of this research was to use ionic crosslinking to improve the mechanical characteristics, absorption of fluid in the wound, and drug release behavior of Cassia alata L. (CA) extract loaded niosomes (CANs) that were incorporated in an alginate-pectin film (A/P). Then, chemically crosslinked A/P hydrogels were obtained by immersing them in different concentrations of calcium chloride (CaCl2) (0.5–1% w/v) for 15–120 s. The degree of crosslinking was controlled by both contact time and CaCl2 concentration. The optimal crosslinking conditions were 1% CaCl2 for 15 seconds. In this study, the following features of the hydrogel films were investigated: physical properties, morphological characteristics, drug loading, in vitro drug release, antibacterial activity, wound healing activity, cytocompatibility profiles, and hemocompatibility. The crosslinked hydrogel films maintained their physical integrity during use, with the 1% film attaining the best results in the shortest period (15 sec). Then, in vitro drug release from the films was examined. Crosslinking was observed to prolong the release of the CA extract from the hydrogel film. Finally, a cell viability experiment was conducted to evaluate the cytotoxicity profile. The A/P composite film exhibited excellent wound dressing qualities and good mechanical properties in preformulation testing. The in vitro drug release profile indicated that the A/P created a regulated drug release profile, and the cell viability experiment revealed that the film was nontoxic and hemocompatible. A/P composite films can be produced using CAN extract as a possible wound dressing. However, further studies in animals and humans are required to determine both safety and effectiveness.
Collapse
|
45
|
Pritzl SD, Morstein J, Kahler S, Konrad DB, Trauner D, Lohmüller T. Postsynthetic Photocontrol of Giant Liposomes via Fusion-Based Photolipid Doping. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:11941-11949. [PMID: 36130117 PMCID: PMC9536078 DOI: 10.1021/acs.langmuir.2c01685] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/06/2022] [Indexed: 06/15/2023]
Abstract
We report on photolipid doping of giant unilamellar vesicles (GUVs) via vesicle fusion with small unilamellar photolipid vesicles (pSUVs), which enables retroactive optical control of the membrane properties. We observe that vesicle fusion is light-dependent, if the phospholipids are neutral. Charge-mediated fusion involving anionic and cationic lipid molecules augments the overall fusion performance and doping efficiency, even in the absence of light exposure. Using phosphatidylcholine analogs with one or two azobenzene photoswitches (azo-PC and dazo-PC) affects domain formation, bending stiffness, and shape of the resulting vesicles in response to irradiation. Moreover, we show that optical membrane control can be extended to long wavelengths using red-absorbing photolipids (red-azo-PC). Combined, our findings present an attractive and practical method for the precise delivery of photolipids, which offers new prospects for the optical control of membrane function.
Collapse
Affiliation(s)
- Stefanie D. Pritzl
- Chair
for Photonics and Optoelectronics, Nano-Institute Munich, Department
of Physics, Ludwig-Maximilians-Universität
(LMU), 80539 Munich, Germany
| | - Johannes Morstein
- Department
of Chemistry, New York University, Silver Center, New York 10003, United States
- Department
of Cellular and Molecular Pharmacology, UCSF, San Francisco, California 94143, United States
| | - Sophia Kahler
- Department
of Chemistry, New York University, Silver Center, New York 10003, United States
| | - David B. Konrad
- Department
of Pharmacy, Ludwig-Maximilians-Universität
(LMU), 81377 Munich, Germany
| | - Dirk Trauner
- Department
of Chemistry, New York University, Silver Center, New York 10003, United States
| | - Theobald Lohmüller
- Chair
for Photonics and Optoelectronics, Nano-Institute Munich, Department
of Physics, Ludwig-Maximilians-Universität
(LMU), 80539 Munich, Germany
| |
Collapse
|
46
|
Hattori Y, Tang M, Torii S, Tomita K, Sagawa A, Inoue N, Yamagishi R, Ozaki KI. Optimal combination of cationic lipid and phospholipid in cationic liposomes for gene knockdown in breast cancer cells and mouse lung using siRNA lipoplexes. Mol Med Rep 2022; 26:253. [PMID: 35686555 PMCID: PMC9218728 DOI: 10.3892/mmr.2022.12769] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/12/2022] [Indexed: 11/05/2022] Open
Abstract
Formulation of cationic liposomes is a key factor that determine the gene knockdown efficiency by cationic liposomes/siRNA complexes (siRNA lipoplexes). Here, to determine the optimal combination of cationic lipid and phospholipid in cationic liposomes for in vitro and in vivo gene knockdown using siRNA lipoplexes, three types of cationic lipid were used, namely 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), dimethyldioctadecylammonium bromide (DDAB) and 11-[(1,3-bis(dodecanoyloxy)-2-((dodecanoyloxy)methyl)propan-2-yl)amino]-N,N,N-trimethyl-11-oxoundecan-1-aminium bromide (TC-1-12). Thereafter, 30 types of cationic liposome composed of each cationic lipid with phosphatidylcholine or phosphatidylethanolamine containing saturated or unsaturated dialkyl chains (C14, C16, or C18) were prepared. The inclusion of phosphatidylethanolamine containing unsaturated and long dialkyl chains with DOTAP- or DDAB-based cationic liposomes induced strong luciferase gene knockdown in human breast cancer MCF-7-Luc cells stably expressing luciferase gene. Furthermore, the inclusion of phosphatidylcholine or phosphatidylethanolamine containing saturated and short dialkyl chains or unsaturated and long dialkyl chains into TC-1-12-based cationic liposomes resulted in high gene knockdown efficacy. When cationic liposomes composed of DDAB/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), TC-1-12/DOPE and TC-1-12/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine were used, significant gene knockdown occurred in the lungs of mice following systemic injection of siRNA lipoplexes. Overall, the present findings indicated that optimal phospholipids in cationic liposome for in vitro and in vivo siRNA transfection were affected by the types of cationic lipid used.
Collapse
Affiliation(s)
- Yoshiyuki Hattori
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| | - Min Tang
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| | - Satomi Torii
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| | - Kana Tomita
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| | - Ayane Sagawa
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| | - Nodoka Inoue
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| | - Reo Yamagishi
- Department of Molecular Pharmaceutics, Hoshi University, Tokyo 142-8501, Japan
| | - Kei-Ichi Ozaki
- Department of Molecular Pathology, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kodo, Kyotanabe, Kyoto 610-0395, Japan
| |
Collapse
|
47
|
Toma I, Porfire AS, Tefas LR, Berindan-Neagoe I, Tomuță I. A Quality by Design Approach in Pharmaceutical Development of Non-Viral Vectors with a Focus on miRNA. Pharmaceutics 2022; 14:1482. [PMID: 35890377 PMCID: PMC9322860 DOI: 10.3390/pharmaceutics14071482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/28/2022] [Accepted: 07/14/2022] [Indexed: 12/10/2022] Open
Abstract
Cancer is the leading cause of death worldwide. Tumors consist of heterogeneous cell populations that have different biological properties. While conventional cancer therapy such as chemotherapy, radiotherapy, and surgery does not target cancer cells specifically, gene therapy is attracting increasing attention as an alternative capable of overcoming these limitations. With the advent of gene therapy, there is increasing interest in developing non-viral vectors for genetic material delivery in cancer therapy. Nanosystems, both organic and inorganic, are the most common non-viral vectors used in gene therapy. The most used organic vectors are polymeric and lipid-based delivery systems. These nanostructures are designed to bind and protect the genetic material, leading to high efficiency, prolonged gene expression, and low toxicity. Quality by Design (QbD) is a step-by-step approach that investigates all the factors that may affect the quality of the final product, leading to efficient pharmaceutical development. This paper aims to provide a new perspective regarding the use of the QbD approach for improving the quality of non-viral vectors for genetic material delivery and their application in cancer therapy.
Collapse
Affiliation(s)
- Ioana Toma
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.T.); (L.R.T.); (I.T.)
| | - Alina Silvia Porfire
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.T.); (L.R.T.); (I.T.)
| | - Lucia Ruxandra Tefas
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.T.); (L.R.T.); (I.T.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania;
| | - Ioan Tomuță
- Department of Pharmaceutical Technology and Biopharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (I.T.); (L.R.T.); (I.T.)
| |
Collapse
|
48
|
Scalzo S, Santos AK, Ferreira HAS, Costa PA, Prazeres PHDM, da Silva NJA, Guimarães LC, E Silva MDM, Rodrigues Alves MTR, Viana CTR, Jesus ICG, Rodrigues AP, Birbrair A, Lobo AO, Frezard F, Mitchell MJ, Guatimosim S, Guimaraes PPG. Ionizable Lipid Nanoparticle-Mediated Delivery of Plasmid DNA in Cardiomyocytes. Int J Nanomedicine 2022; 17:2865-2881. [PMID: 35795081 PMCID: PMC9252585 DOI: 10.2147/ijn.s366962] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/19/2022] [Indexed: 12/18/2022] Open
Abstract
Introduction Gene therapy is a promising approach to be applied in cardiac regeneration after myocardial infarction and gene correction for inherited cardiomyopathies. However, cardiomyocytes are crucial cell types that are considered hard-to-transfect. The entrapment of nucleic acids in non-viral vectors, such as lipid nanoparticles (LNPs), is an attractive approach for safe and effective delivery. Methods Here, a mini-library of engineered LNPs was developed for pDNA delivery in cardiomyocytes. LNPs were characterized and screened for pDNA delivery in cardiomyocytes and identified a lead LNP formulation with enhanced transfection efficiency. Results By varying lipid molar ratios, the LNP formulation was optimized to deliver pDNA in cardiomyocytes with enhanced gene expression in vitro and in vivo, with negligible toxicity. In vitro, our lead LNP was able to reach a gene expression greater than 80%. The in vivo treatment with lead LNPs induced a twofold increase in GFP expression in heart tissue compared to control. In addition, levels of circulating myeloid cells and inflammatory cytokines remained without significant changes in the heart after LNP treatment. It was also demonstrated that cardiac cell function was not affected after LNP treatment. Conclusion Collectively, our results highlight the potential of LNPs as an efficient delivery vector for pDNA to cardiomyocytes. This study suggests that LNPs hold promise to improve gene therapy for treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Sérgio Scalzo
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anderson K Santos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Heloísa A S Ferreira
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro A Costa
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro H D M Prazeres
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natália J A da Silva
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lays C Guimarães
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mário de Morais E Silva
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marco T R Rodrigues Alves
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso T R Viana
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Itamar C G Jesus
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alice P Rodrigues
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anderson O Lobo
- Department of Materials Engineering, Federal University of Piauí, Teresina, PI, Brazil
| | - Frederic Frezard
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | |
Collapse
|
49
|
Gómez-Aguado I, Rodríguez-Castejón J, Beraza-Millor M, Rodríguez-Gascón A, Del Pozo-Rodríguez A, Solinís MÁ. mRNA delivery technologies: Toward clinical translation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 372:207-293. [PMID: 36064265 DOI: 10.1016/bs.ircmb.2022.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Messenger RNA (mRNA)-therapies have recently taken a huge step toward clinic thanks to the first mRNA-based medicinal products marketed. mRNA features for clinical purposes are improved by chemical modifications, but the inclusion in a delivery system is a regular requirement. mRNA nanomedicines must be designed for the specific therapeutic purpose, protecting the nucleic acid and facilitating the overcoming of biological barriers. Polymers, polypeptides, and cationic lipids are the main used materials to design mRNA delivery systems. Among them, lipid nanoparticles (LNPs) are the most advanced ones, and currently they are at the forefront of preclinical and clinical evaluation in several fields, including immunotherapy (against infectious diseases and cancer), protein replacement, gene editing and regenerative medicine. This chapter includes an overview on mRNA delivery technologies, with special interest in LNPs, and the most recent advances in their clinical application. Liposomes are the mRNA delivery technology with the highest clinical translation among LNPs, whereas the first clinical trial of a therapeutic mRNA formulated in exosomes has been recently approved for protein replacement therapy. The first mRNA products approved by the regulatory agencies worldwide are LNP-based mRNA vaccines against viral infections, specifically against the 2019 coronavirus disease (COVID-19). The clinical translation of mRNA-therapies for cancer is mainly focused on three strategies: anti-cancer vaccination by means of delivering cancer antigens or acting as an adjuvant, mRNA-engineered chimeric antigen receptors (CARs) and T-cell receptors (TCRs), and expression of antibodies and immunomodulators. Cancer immunotherapy and, more recently, COVID-19 vaccines spearhead the advance of mRNA clinical use.
Collapse
Affiliation(s)
- Itziar Gómez-Aguado
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Julen Rodríguez-Castejón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Marina Beraza-Millor
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - María Ángeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain.
| |
Collapse
|
50
|
Masud AA, Alsharif FM, Creameans JW, Perdeh J, Feola DJ, Venditto VJ. Optimization and Characterization of a Liposomal Azithromycin Formulation for Alternative Macrophage Activation. FRONTIERS IN DRUG DELIVERY 2022; 2:908709. [PMID: 36407498 PMCID: PMC9670256 DOI: 10.3389/fddev.2022.908709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Liposomal azithromycin (L-AZM) promotes macrophage polarization toward an M2-like phenotype in the context of myocardial infarction that results in improved cardiovascular outcomes in mice. To improve upon this formulation, we sought to identify optimized formulation, stability, and biological activity parameters necessary to enhance the immunomodulatory activity and efficacy of L-AZM. While our parent formulation contains a mixture of long-chain saturated phosphatidylcholine and phosphatidylglycerol lipids, we evaluated a series of formulations with different amounts of unsaturated lipids and cholesterol with the goal of improving the loading capacity and stability of the formulations. We also introduce fusogenic lipids to improve the cytosolic delivery to enhance the immune modulatory properties of the drug. To achieve these goals, we initially prepared a library of 24 formulations using thin film hydration and assessed the resultant liposomes for size and polydispersity. Five lead formulations were identified based on low polydispersity (<0.3) and stability over time. The lead formulations were then evaluated for stability in serum using dialysis and macrophage polarization activity in vitro as measured by decreased IL-12 expression. Collectively, our data indicate that the formulation components drive the balance between encapsulation efficiency and stability and that all the lead liposomal formulations improve in vitro alternative macrophage activation as compared to free AZM.
Collapse
Affiliation(s)
- Abdullah A. Masud
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Fahd M. Alsharif
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Jarrod W. Creameans
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Jasmine Perdeh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - David J. Feola
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Vincent J. Venditto
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| |
Collapse
|