1
|
Lynn AY, Shin K, Eaton DA, Rose M, Zhang X, Ene M, Grundler J, Deschenes E, Rivero R, Bracaglia LG, Glazer PM, Stitelman DH, Saltzman WM. Investigation of the protein corona and biodistribution profile of polymeric nanoparticles for intra-amniotic delivery. Biomaterials 2025; 320:123238. [PMID: 40064138 PMCID: PMC11972154 DOI: 10.1016/j.biomaterials.2025.123238] [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: 11/18/2024] [Revised: 02/03/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
When exposed to the biological environment, nanoparticles (NPs) form a protein corona that influences delivery profile. We present a study of protein corona formation and NP biodistribution in amniotic fluid (AF) for poly(lactic-co-glycolic acid) (PLGA) and poly(lactic-acid) (PLA) NPs, with and without polyethylene glycol (PEG), as well as poly(amine-co-ester)-PEG (PACE-PEG) NPs. The presence of surface PEG and polyvinyl alcohol (PVA) were characterized to investigate surfactant role in determining protein corona formation. The surface density of PEG groups demonstrated an inverse correlation with the total amount of protein surface adsorption. All PEGylated NPs exhibited a dense brush conformation and demonstrated higher levels of stability in AF than non-PEGylated NPs. The protein corona composition varied by core polymer, while the amount of protein adsorption varied by PEGylation status. In A549 cells, in vitro cellular association of each NP type correlated with the amount of albumin that was found in the protein corona. In vivo, only PEGylated NPs were able successfully distribute to fetal organs, likely due to the enhanced stability imparted by PEG. PLGA-PEG and PACE-PEG NPs had both high levels of albumin in the protein corona and high biodistribution to the fetal lung, consistent with the association with lung cells in vitro. PLA-PEG NPs distributed exclusively to the fetal bowel, which we propose is associated with known gastrointestinal targeting keratin proteins. By furthering our understanding of polymeric NP behavior in AF, this novel study provides a basis for optimization of intra-amniotic NP delivery systems targeting congenital pulmonary and gastrointestinal diseases.
Collapse
Affiliation(s)
- Anna Y Lynn
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA
| | - Kwangsoo Shin
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA
| | - David A Eaton
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA
| | - Micky Rose
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA
| | - Xianzhi Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA
| | - Madalina Ene
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA
| | - Julian Grundler
- Department of Chemistry, Yale University, New Haven, CT, 06510, USA
| | - Emily Deschenes
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA
| | - Rachel Rivero
- Department of Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Laura G Bracaglia
- Department of Chemical and Biological Engineering, Villanova University, Villanova, PA, 19085, USA
| | - Peter M Glazer
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - David H Stitelman
- Department of Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06510, USA; Department of Chemical & Environmental Engineering, Yale University, New Haven, CT, 06510, USA; Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, 06510, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
2
|
Tian H, Deng H, Liu X, Liu C, Zhang C, Leong KW, Fan X, Ruan J. A novel FTO-targeting nanodrug induces disulfidptosis and ameliorates the suppressive tumor immune environment to treat uveal melanoma. Biomaterials 2025; 319:123168. [PMID: 40015005 DOI: 10.1016/j.biomaterials.2025.123168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Uveal melanoma (UM) is the most prevalent primary ocular malignancy in adults, with high lethality and limited effective treatment options. Despite identified driver mutations in GNAQ, GNA11, and BAP1, therapeutic advancements have been minimal. This study highlights the pivotal role of N6-methyladenosine (m6A) modifications in UM pathogenesis and progression, focusing on the demethylase FTO as a therapeutic target. Elevated FTO expression in UM tissues correlates with decreased m6A levels, increased aggressiveness, and poor prognosis. The FTO inhibitor meclofenamic acid (MA) restored m6A levels, upregulated SLC7A11, and induced disulfidptosis, a unique form of cell death triggered by GSH depletion and NADPH consumption. To address MA's limitations in bioavailability and tumor targeting, we developed an MA-loaded nucleic acid nanodrug (SNAMA). SNAMA demonstrated effective tumor growth inhibition in orthotopic and metastatic UM models through GSH-responsive release and m6A-mediated disulfidptosis activation. Incorporating a PD-L1 aptamer into SNAMA further improved tumor targeting and immune modulation, enhancing therapeutic efficacy. This study identifies FTO as a critical target for UM therapy and introduces SNAMA-apt as a promising nanodrug. The findings offer a foundation for m6A-targeted approaches in UM and other malignancies, addressing bioavailability, targeting, and immune evasion challenges.
Collapse
Affiliation(s)
- Hao Tian
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China; Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Hongpei Deng
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China
| | - Xinlong Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Chang Liu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China.
| | - Jing Ruan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China; Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
3
|
Lee K, Seong HG, Russell TP, Emrick T. Thiol- and Disulfide-Functionalized Polycyclooctene: Metathesis Polymerization, Degradation, and Reformation. ACS Macro Lett 2025; 14:616-621. [PMID: 40314497 DOI: 10.1021/acsmacrolett.5c00245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
The enormous global production and use of polymers and plastics, combined with slower and less efficient disposal and recycling methods, have led to a worldwide growth in plastic waste. Here, we describe an approach that builds degradability into macromolecular polyolefins through the design of telechelic oligomers with reactive functional groups that enables control over polymer deconstruction and reconstruction. Our work exploits disulfide-containing polymers that have emerged as promising cyclable materials due to their dynamic reversibility (bond formation and cleavage) driven by redox chemistry under mild conditions. Specifically, we describe the synthesis of a telechelic α,ω-dithiopolycyclooctene (PCOE) by ring-opening metathesis polymerization using a dithioacetate chain transfer agent, followed by deprotection to convert the chain-end thioacetates to thiols. This process was studied towards control over the degree of oxidation to yield disulfide-containing PCOE, followed by an evaluation of reductive degradation and oxidative repolymerization. Overall, this approach facilitates the production of disulfide-containing unsaturated polyolefins and the integration of degradable and reformable moieties into soluble, processable, metathesis-derived polymers.
Collapse
Affiliation(s)
- Kyoungwon Lee
- Polymer Science & Engineering Department, Conte Center for Polymer Research, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Hong-Gyu Seong
- Polymer Science & Engineering Department, Conte Center for Polymer Research, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Thomas P Russell
- Polymer Science & Engineering Department, Conte Center for Polymer Research, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Todd Emrick
- Polymer Science & Engineering Department, Conte Center for Polymer Research, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
4
|
Badal AK, Nayek A, Dhar R, Karmakar S. MicroRNA nanoformulation: a promising approach to anti-tumour activity. Invest New Drugs 2025:10.1007/s10637-025-01534-7. [PMID: 40366533 DOI: 10.1007/s10637-025-01534-7] [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: 01/30/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025]
Abstract
Cancer is a major cause of morbidity and mortality, making it one of the most debilitating diseases in our time. Despite advancements in therapeutic strategies, the development of chemoresistance and the occurrence of secondary tumours pose significant challenges. While several promising anti-tumour agents have been identified, their clinical utility is often limited due to toxicity and associated side effects. MicroRNAs (mi-RNAs) are critical regulators of gene expression, and their altered levels are closely linked to cancer development and progression. Although some microRNAs have shown potential as biomarkers for cancer detection, their integration into routine clinical practice has yet to be realized. Numerous candidate microRNAs exhibit therapeutic potential for cancer treatment; however, further research is needed to create efficient, patient-compliant, and customized drug delivery systems. In recent decades, various nanotechnology platforms have successfully transitioned to clinical trials, particularly in the field of RNA nanotechnology. Several RNA nanoparticles have been developed to address key challenges in vivo for targeting cancer, demonstrating favourable biodistribution characteristics. Studies have shown that RNA nanoparticles, characterized by precise stoichiometry and homogeneity, can effectively target tumour cells while avoiding aggregation in normal, healthy tissues following systemic injection. Animal models have demonstrated that RNA nanoparticles can deliver therapeutics such as siRNA and anti-microRNA, effectively inhibiting tumour growth. Using nanoparticles conjugated with antibodies and/or peptides enhances the targeted delivery and sustained release of microRNAs and anti-microRNAs, which may reduce the required therapeutic dosage and minimize systemic and cellular damage. This review focuses on developing microRNA nanoformulations to improve cellular uptake, bioavailability, and accumulation at tumour sites, assessing their potential anti-tumour efficacy against various types of malignancies. The significance of these advancements in clinical oncology cannot be overstated.
Collapse
Affiliation(s)
| | - Arnab Nayek
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Ruby Dhar
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India.
| | - Subhradip Karmakar
- All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India.
| |
Collapse
|
5
|
Dai Y, Yang L, Cao G, Mo L, Yang C, Zhu Y, Guo Y, Hong Y, Xu H, Lu S, Du S, He J. Combination therapy and drug co-delivery systems for atherosclerosis. J Control Release 2025; 381:113543. [PMID: 39986476 DOI: 10.1016/j.jconrel.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of plaque within the arteries. Despite advances in therapeutic strategies including anti-inflammatory, antioxidant, and lipid metabolism modulation treatments over the past two decades, the treatment of atherosclerosis remains challenging, as arterial damage is the result of interconnected pathological factors. Therefore, current monotherapies often fail to address the complex nature of this disease, leading to insufficient therapeutic outcomes. This review addressed this paucity of effective treatment options by comprehensively exploring the potential for combination therapies and advanced drug co-delivery systems for the treatment of atherosclerosis. We investigated the pathological features of and risk factors for atherosclerosis, underscoring the importance of drug combination therapies for the treatment of atherosclerotic diseases. We discuss herein mathematical models for quantifying the efficacy of the combination therapies and provide a systematic summary of drug combinations for the treatment of atherosclerosis. We also provide a detailed review of the latest advances in nanoparticle-based drug co-delivery systems for the treatment of atherosclerosis, focusing on the design of carriers with high biocompatibility and efficacy. By exploring the possibilities and challenges inherent to this approach, we aim to highlight cutting-edge technologies that can foster the development of innovative strategies, optimize drug co-administration, improve treatment outcomes, and reduce the burden of atherosclerosis-related morbidity and mortality on the healthcare system.
Collapse
Affiliation(s)
- Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Guosheng Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Hanlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| |
Collapse
|
6
|
Komatsu S, Suzuki T, Kosukegawa Y, Kawase M, Matsuyama T, Asoh TA, Kikuchi A. Preparation of thermoresponsive core-corona particles for controlled phagocytosis via surface properties and particle shape transformation. J Control Release 2025; 381:113652. [PMID: 40120692 DOI: 10.1016/j.jconrel.2025.113652] [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: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Cell-particle interactions, such as phagocytosis, exhibit variability based on particle shape, surface physical properties, and diameter. These interactions can be intentionally modified through in situ change in the physical characteristics of the particulate materials. By manipulating both the surface properties and shape of the particles, it may be feasible to regulate their interactions with cells. Objective of this research is to prepare thermoresponsive core-corona particles those undergo transformation and alteration in surface solubility near physiological temperature and to investigate particle shape- and surface physical property-dependent phagocytosis. The glass transition temperature of the prepared particles was controlled via the composition of the polymer core. Rod-type particles, prepared by uniaxially stretching particle-containing films at above the glass transition temperature of the core-forming materials, demonstrated reduced phagocytosis by macrophages compared to that of spherical particles. Furthermore, the physical properties of the particle surface exerted a significant influence on phagocytosis, with hydrophobic particles being more readily engulfed. Consequently, precise control of phagocytosis can be controlled by manipulating the particle's shape and surface properties. The prepared particles have potential applications as drug delivery system carriers, enabling the regulation of cell interactions via particle shape and surface physical properties induced by temperature changes.
Collapse
Affiliation(s)
- Syuuhei Komatsu
- Department of Materials Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan
| | - Takuma Suzuki
- Department of Materials Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan
| | - Yota Kosukegawa
- Department of Materials Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan
| | - Masatoshi Kawase
- Department of Materials Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan
| | - Takuya Matsuyama
- Department of Materials Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan
| | - Taka-Aki Asoh
- Department of Materials Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan
| | - Akihiko Kikuchi
- Department of Materials Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika, Tokyo 125-8585, Japan.
| |
Collapse
|
7
|
Abe S, Takata H, Shimizu T, Kawaguchi Y, Fukuda S, Yamamoto H, Ando H, Ishida T. Impact of pre-existing anti-polyethylene glycol (PEG) IgM on biodistribution and humoral response of intramuscularly administered PEGylated mRNA loaded lipid nanoparticle. J Control Release 2025; 383:113821. [PMID: 40339658 DOI: 10.1016/j.jconrel.2025.113821] [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: 01/03/2025] [Revised: 05/02/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
With the approval of mRNA vaccines for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), lipid nanoparticles (LNP) have emerged as a powerful tool for nucleic acid delivery. Modification of LNP with polyethylene glycol (PEG)-lipids contributes to their in vitro and in vivo stability by reducing aggregation of the particles. Despite the general acknowledgement of the low immunogenicity of PEG, there are numerous reports of the occurrence of anti-PEG antibodies in the blood of healthy individuals. Furthermore, there are reports that pre-existing anti-PEG IgM antibodies attenuated the efficacy of PEG-modified drugs administered systemically. Skeletal muscles, the administration site for mRNA vaccines, are highly vascularized by a network of blood vessels to provide them with nutrients and oxygen. Since skeletal muscles can contain circulating anti-PEG antibodies or the administered mRNA-LNP extravasate into bloodstream, the purpose of this study was to evaluate the effects of pre-existing anti-PEG IgM on the protein translation, biodistribution, and humoral responses to mRNA-loaded LNP (mRNA-LNP) administered intramuscularly (i.m.) in mice. We found that the presence of anti-PEG IgM in blood has only a minor effect on the translation and distribution of mRNA-LNP in the localized muscle area where the mRNA-LNP were administered. Circulating anti-PEG IgM did not increase the total accumulation of mRNA-LNP in the liver, but did markedly diminish its protein translation because the mRNA-LNP were delivered primarily to Kupffer cells rather than to hepatocytes. Binding of anti-PEG IgM to mRNA-LNP, and subsequent complement activation, suppressed mRNA translation loaded in LNP in the liver. Repeated intramuscular injections of SARS-CoV-2 Spike protein mRNA-LNP elicited a robust immune response. Our results indicate that the presence of circulating anti-PEG IgM does not interfere with the efficiency of mRNA vaccines administered i.m. in mice. We can speculate that non-specific translation of mRNA vaccines in somatic cells may be inhibited in vivo by circulating anti-PEG IgM, reducing adverse effects such as hepatitis and myocarditis that are caused by immune responses against translated antigen proteins.
Collapse
Affiliation(s)
- Shunji Abe
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505 Tokushima, Japan
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Yoshino Kawaguchi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Shoichiro Fukuda
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Haruka Yamamoto
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505 Tokushima, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, 1-78-1, Sho-machi, Tokushima 770-8505, Japan; Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, 770-8505 Tokushima, Japan.
| |
Collapse
|
8
|
Del Bene A, D'Aniello A, Mottola S, Mazzarella V, Cutolo R, Campagna E, Benedetti R, Altucci L, Cosconati S, Di Maro S, Messere A. From genetic code to global health: the impact of nucleic acid vaccines on disease prevention and treatment. RSC Med Chem 2025:d5md00032g. [PMID: 40337306 PMCID: PMC12053015 DOI: 10.1039/d5md00032g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/19/2025] [Indexed: 05/09/2025] Open
Abstract
Vaccinology has revolutionized modern medicine, delivering groundbreaking solutions to prevent and control infectious diseases while pioneering innovative strategies to tackle non-infectious challenges, including cancer. Traditional vaccines faced inherent limitations, driving the evolution of next-generation vaccines such as subunit vaccines, peptide-based vaccines, and nucleic acid-based platforms. Among these, nucleic acid-based vaccines, including DNA and mRNA technologies, represent a major innovation. Pioneering studies in the 1990s demonstrated their ability to elicit immune responses by encoding specific antigens. Recent advancements in delivery systems and molecular engineering have overcome initial challenges, enabling their rapid development and clinical success. This review explores nucleic acid-based vaccines, including chemically modified variants, by examining their mechanisms, structural features, and therapeutic potential, while underscoring their pivotal role in modern immunization strategies and expanding applications across contemporary medicine.
Collapse
Affiliation(s)
- Alessandra Del Bene
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | | | - Salvatore Mottola
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Vincenzo Mazzarella
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Roberto Cutolo
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Erica Campagna
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Vico Luigi De Crecchio 1 80138 Naples Italy
- Program of Medical Epigenetics, Vanvitelli Hospital 80138 Naples Italy
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Vico Luigi De Crecchio 1 80138 Naples Italy
- Program of Medical Epigenetics, Vanvitelli Hospital 80138 Naples Italy
- Biogem Institute of Molecular and Genetic Biology 83031 Ariano Irpino Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli" Vico Luigi De Crecchio 1 80138 Naples Italy
| | - Sandro Cosconati
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Salvatore Di Maro
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| | - Anna Messere
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania "Luigi Vanvitelli" Caserta Italy
| |
Collapse
|
9
|
Liu Y, Biesel A, Kamal MAM, Latta L, Loretz B, Hirsch AKH, Lee S, Lehr CM. Tobramycin crosslinking improves the colloidal stability of arginine chitosan biodynamers for safe and efficient siRNA delivery. Int J Biol Macromol 2025; 311:143420. [PMID: 40274164 DOI: 10.1016/j.ijbiomac.2025.143420] [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: 02/16/2025] [Revised: 04/09/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
We previously reported proteoid biodynamers to form dynamic nanocomplexes (dynaplexes, DPs) with siRNA and their efficient transfection via dynamic responsive changes in endosomal environments. However, their limited colloidal stability requires chemical crosslinking, which may cause toxicity. To address these limitations, we designed biodynamers using positively charged aldehyde chitosan (ACh) as the backbone, which was functionalized with arginine hydrazide (Arg) and/or tobramycin (Tob) to create either Arg-ACh, Tob-ACh or Arg/Tob-ACh, respectively. While the additional positive charges introduced by Arg improved the siRNA complexation, the multiple amine groups in Tob acted as internal crosslinkers. Additionally, the resulting siRNA-loaded DPs were coated with hyaluronic acid (HA), which further enhanced colloidal stability. Compared to proteoid biodynamers, these novel functionalized chitosan biodynamers provided adequate gene silencing efficiency without the need for potentially harmful additional crosslinkers.
Collapse
Affiliation(s)
- Yun Liu
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; Department of Pharmacy, Saarland University, PharmaScienceHub (PSH), Saarbrücken, Germany
| | - Achim Biesel
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; Department of Pharmacy, Saarland University, PharmaScienceHub (PSH), Saarbrücken, Germany
| | - Mohamed A M Kamal
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; Department of Pharmacy, Saarland University, PharmaScienceHub (PSH), Saarbrücken, Germany
| | - Lorenz Latta
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Brigitta Loretz
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Anna K H Hirsch
- Department of Pharmacy, Saarland University, PharmaScienceHub (PSH), Saarbrücken, Germany; Department of Drug Design and Optimisation, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Sangeun Lee
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; Department of Pharmacy, Saarland University, PharmaScienceHub (PSH), Saarbrücken, Germany.
| | - Claus-Michael Lehr
- Department of Drug Delivery Across Biological Barriers, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; Department of Pharmacy, Saarland University, PharmaScienceHub (PSH), Saarbrücken, Germany.
| |
Collapse
|
10
|
Linderman SW, DeRidder L, Sanjurjo L, Foote MB, Alonso MJ, Kirtane AR, Langer R, Traverso G. Enhancing immunotherapy with tumour-responsive nanomaterials. Nat Rev Clin Oncol 2025; 22:262-282. [PMID: 40050505 DOI: 10.1038/s41571-025-01000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
The targeted delivery of immunotherapies to tumours using tumour-responsive nanomaterials is a promising area of cancer research with the potential to address the limitations of systemic administration such as on-target off-tumour toxicities and a lack of activity owing to the immunosuppressive tumour microenvironment (TME). Attempts to address these challenges include the design and functionalization of nanomaterials capable of releasing their cargoes in response to specific TME characteristics, thus facilitating the targeted delivery of immune-checkpoint inhibitors, cytokines, mRNAs, vaccines and, potentially, chimaeric antigen receptors as well as of agents that modulate the extracellular matrix and induce immunogenic cell death. In this Review, we describe these various research efforts in the context of the dynamic properties of the TME, such as pH, reductive conditions, reactive oxygen species, hypoxia, specific enzymes, high levels of ATP and locoregional aspects, which can be leveraged to enhance the specificity and efficacy of nanomaterial-based immunotherapies. Highlighting preclinical successes and ongoing clinical trials, we evaluate the current landscape and potential of these innovative approaches. We also consider future research directions as well as the most important barriers to successful clinical translation, emphasizing the transformative potential of tumour-responsive nanomaterials in overcoming the barriers that limit the activity of traditional immunotherapies, thus improving patient outcomes.
Collapse
Affiliation(s)
- Stephen W Linderman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Hospital Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louis DeRidder
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- IMDEA Nanosciences Institute, Madrid, Spain
| | - Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Langer
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
11
|
Wang B, Zhang S, Guo Y, Gao W, Wu H, Wang J, Wang Y, Tang C, Liu L. CBX2 as a therapeutic target in colorectal cancer: insights into the altered chromatin accessibility via RUNX1-CBX2-MAP4K1 axis. Oncogene 2025; 44:909-926. [PMID: 40082555 DOI: 10.1038/s41388-025-03331-1] [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: 10/25/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
Chromobox homolog 2 (CBX2), a component of the polycomb repressive complex 1, is overexpressed in various cancers, but its specific role in colorectal cancer (CRC) is not fully understood. This study aimed to characterize the functional and regulatory roles of CBX2 in CRC. Tissue microarray analysis revealed the elevated CBX2 levels in tumor compared to adjacent normal tissues, which is significantly correlated with poor prognosis. Gain and loss of function studies demonstrated that CBX2 significantly promoted CRC progression and chemoresistance in cell lines, patient-derived CRC organoids and xenografts. In the AOM/DSS mouse model, treatment with the innovatively-developed cy5-PBAE/siCBX2 nanoparticle significantly reduced tumor aggressiveness. Mechanistic studies unveiled that the transcription factor RUNX1 is the positive regulator of CBX2. RNA-seq, ATAC-seq and CUT & RUN results indicated CBX2 knockdown induced epigenetic changes, especially alterations in chromatin accessibility. Moreover, we further identified MAP4K1 as a target gene of RUNX1-CBX2, with significant clinical and prognostic relevance in CRC. Collectively, these findings suggest the pivotal role of RUNX1-CBX2-MAP4K1 axis in CRC progression and underscore CBX2 as a promising biomarker and therapeutic target. The regulatory function of CBX2 on chromatin accessibility and the role of the RUNX1-CBX2-MAP4K1-pERK axis in the progression of colorectal cancer.
Collapse
Affiliation(s)
- Bangting Wang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China
- The Friendship Hospital of Ili Kazkh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Yining, China
| | - Shijie Zhang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Yumeng Guo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wenqing Gao
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Hao Wu
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Jiankun Wang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Yan Wang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China.
- The Friendship Hospital of Ili Kazkh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Yining, China.
| | - Chunming Tang
- College of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Li Liu
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China.
| |
Collapse
|
12
|
Neupane R, Malla S, Karthikeyan C, Asbhy CR, Boddu SHS, Jayachandra Babu R, Tiwari AK. Endocytic highways: Navigating macropinocytosis and other endocytic routes for precision drug delivery. Int J Pharm 2025; 673:125356. [PMID: 39956408 DOI: 10.1016/j.ijpharm.2025.125356] [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: 09/15/2024] [Revised: 12/22/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Drug molecules can reach intracellular targets by different mechanisms, such as passive diffusion, active transport, and endocytosis. Endocytosis is the process by which cells engulf extracellular material by forming a vesicle and transporting it into the cells. In addition to its biological functions, endocytosis plays a vital role in the internalization of the therapeutic molecules. Clathrin-mediated endocytosis, caveolar endocytosis, and macropinocytosis are the most researched routes in the field of drug delivery. In addition to conventional small therapeutic molecules, the use of nanoformulations and large molecules, such as nucleic acids, peptides, and antibodies, have broadened the field of drug delivery. Although the majority of small therapeutic molecules can enter cells via passive diffusion, large molecules, and advanced targeted delivery systems, such as nanoparticles, are internalized by the endocytic route. Therefore, it is imperative to understand the characteristics of the endocytic routes in greater detail to design therapeutic molecules or formulations for successful delivery to the intracellular targets. This review highlights the prospects and limitations of the major endocytic routes for drug delivery, with a major emphasis on macropinocytosis. Since macropinocytosis is a non-selective uptake of extracellular matrix, the selective induction of macropinocytosis, using compounds that induce macropinocytosis and modulate macropinosome trafficking pathways, could be a potential approach for the intracellular delivery of diverse therapeutic modalities. Furthermore, we have summarized the characteristics associated with the formulations or drug carriers that can affect the endocytic routes for cellular internalization. The techniques that are used to study the intracellular uptake processes of therapeutic molecules are briefly discussed. Finally, the major limitations for intracellular targeting, endo-lysosomal degradation, and different approaches that have been used in overcoming these limitations, are highlighted in this review.
Collapse
Affiliation(s)
- Rabin Neupane
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA
| | - Chandrabose Karthikeyan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak 484887, India
| | - Charles R Asbhy
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY 10049, USA
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, AL 36849, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
13
|
Park J, Rhoo KY, Kim Y, Kim YS, Paik SR. Cell-Division-Independent Rapid Expression of DNA Delivered with α-Synuclein-Gold Nanoparticle Conjugates. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14846-14858. [PMID: 40014054 DOI: 10.1021/acsami.4c17967] [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: 02/28/2025]
Abstract
Gene delivery is a primary technology employed in diverse areas of biomedical science, from gene therapy to gene editing, cancer treatment, and stem cell research. Here, we introduce a gene delivery system utilizing an intrinsically disordered protein of α-synuclein (αS) demonstrated to interact with lipid membranes by transforming its original random structure to an α-helix. Since the helix bundle formation is a signature of cell-penetrating peptides for membrane translocation, a multitude of αS(Y136C)s replacing tyrosine at the C-terminus with cysteine were covalently attached onto gold nanoparticles (AuNPs) in a specific orientation with the helix-forming basic N-termini exposed outward. The resulting αS(Y136C)-AuNP conjugates were found to exhibit a rapid gene expression without causing cytotoxicity when the gene of the enhanced green fluorescent protein (EGFP) was delivered with the conjugates into the cells. Based on inhibition studies toward endocytosis and mitosis, the αS(Y136C)-AuNP/DNA complex was demonstrated to take both endosomal and non-endosomal intracellular transport pathways. The DNA translocation into the nucleus was independent of cell division. This nondisruptive and rapid DNA transfection by αS(Y136C)-AuNPs allowed a successful delivery of granzyme A gene leading to cellular pyroptosis. Modifications of αS(Y136C)-AuNP/DNA complex, such as antibody immobilization and replacement of DNA with biological suprastructures including RNA, protein, and nonbiological fusion materials, would allow the intracellular delivery system to be applied in diverse areas of future biotechnology.
Collapse
Affiliation(s)
- Jeongha Park
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Kun Yil Rhoo
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yunsoo Kim
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Sik Kim
- Department of Pathology, Ansan Hospital, Korea University College of Medicine, Ansan 15355, Republic of Korea
| | - Seung R Paik
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
14
|
Li H, Ma L, Zhu N, Liang X, Tian X, Liu K, Fu X, Wang X, Zhang H, Chen H, Liu Q, Yang J. Mesenchymal stromal cells surface engineering for efficient hematopoietic reconstitution. Biomaterials 2025; 314:122882. [PMID: 39423513 DOI: 10.1016/j.biomaterials.2024.122882] [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: 06/19/2024] [Revised: 09/20/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Mesenchymal stromal cells (MSCs) are believed to migrate to injury sites, release chemical attractants, and either recruit local stem cells or modulate the immune system positively. Although MSCs are highly desired for their potential to reduce inflammation and promote tissue regeneration, their limited lifespan restricts their applications. This study presents a simple approach for protecting MSCs with epigallocatechin-3-gallate (EGCG) and magnesium (Mg) based metal-organic framework coatings (E-Mg@MSC). The layer strengthens MSCs resistant to harmful stresses and creates a favorable microenvironment for repair by providing Mg to facilitate MSCs' osteogenic differentiation and using EGCG to neutralize excessive reactive oxygen species (ROS). E-Mg@MSC serves as a treatment for hematopoietic injury induced by ionizing radiation (IR). Coated MSCs exhibit sustained secretion of hematopoietic growth factors and precise homing to radiation-sensitive tissues. In vivo studies show substantial enhancement in hematopoietic system recovery and multi-organ protection. Mechanistic investigations suggest that E-Mg@MSC mitigates IR-induced ROS, cell apoptosis, and ferroptosis, contributing to reduced radiation damage. The system represents a versatile and compelling strategy for cell-surface engineering with functional materials to advance MSCs therapy.
Collapse
Affiliation(s)
- Huiyang Li
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Lifei Ma
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Ni Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Xiaoyu Liang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Xinxin Tian
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Kaijing Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Xue Fu
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Xiaoli Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China
| | - Hailing Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China.
| | - Houzao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300192, China.
| | - Jing Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China.
| |
Collapse
|
15
|
Bolideei M, Barzigar R, Gahrouei RB, Mohebbi E, Haider KH, Paul S, Paul MK, Mehran MJ. Applications of Gene Editing and Nanotechnology in Stem Cell-Based Therapies for Human Diseases. Stem Cell Rev Rep 2025:10.1007/s12015-025-10857-0. [PMID: 40014250 DOI: 10.1007/s12015-025-10857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Stem cell research is a dynamic and fast-advancing discipline with great promise for the treatment of diverse human disorders. The incorporation of gene editing technologies, including ZFNs, TALENs, and the CRISPR/Cas system, in conjunction with progress in nanotechnology, is fundamentally transforming stem cell therapy and research. These innovations not only provide a glimmer of optimism for patients and healthcare practitioners but also possess the capacity to radically reshape medical treatment paradigms. Gene editing and nanotechnology synergistically enhance stem cell-based therapies' precision, efficiency, and applicability, offering transformative potential for treating complex diseases and advancing regenerative medicine. Nevertheless, it is important to acknowledge that these technologies also give rise to ethical considerations and possible hazards, such as inadvertent genetic modifications and the development of genetically modified organisms, therefore creating a new age of designer infants. This review emphasizes the crucial significance of gene editing technologies and nanotechnology in the progress of stem cell treatments, particularly for degenerative pathologies and injuries. It emphasizes their capacity to restructure and comprehensively revolutionize medical treatment paradigms, providing fresh hope and optimism for patients and healthcare practitioners.
Collapse
Affiliation(s)
- Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Rambod Barzigar
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India
| | - Razieh Bahrami Gahrouei
- Department of Pharmacy PES College, Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka, India
| | - Elham Mohebbi
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois School of Medicine, Springfield, IL, USA
| | - Khawaja Husnain Haider
- Sulaiman AlRajhi Medical School, Al Bukayriyah, AlQaseem, 52726, Kingdom of Saudi Arabia
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA.
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Mohammad Javad Mehran
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India.
| |
Collapse
|
16
|
Lu X, Xia H, Gao W, Pan X, Yan Y, Zhang R, He Y, Wang Y, Chen B, Mei D. A pH-Responsive and Guanidinium-Rich Nanoadjuvant Efficiently Delivers STING Agonist for Cancer Immunotherapy. ACS NANO 2025; 19:6758-6770. [PMID: 39933120 DOI: 10.1021/acsnano.4c10202] [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: 02/13/2025]
Abstract
As natural agonists of the stimulator of interferon genes (STING), cyclic dinucleotides (CDNs) have been identified as promising immunotherapies that trigger a potent immune response against tumors. However, the low stability, rapid clearance, inadequate cellular uptake, and inefficient cytosol localization heavily hinder the therapeutic efficacy of the hydrophilic and negatively charged 2', 3'-cyclic-GMP-AMP (cGAMP). How to efficiently deliver cGAMP into the endoplasmic reticulum (ER) to activate STING for immune priming remains challenging. Here, we report a pH-responsive and guanidinium-rich STING nanoagonist (nPGSA) for cytosol delivery of cGAMP. Compared with free cGAMP, nPGSA achieves up to a 37.4-fold enhancement of cellular internalization. The pH-sensitive and guanidinium-functional design facilitates quick release and endosome escape, thus enabling precise ER targeting of cGAMP and 33.9-fold amplification of STING sensibilization. Furthermore, through the modulation of tumor-associated macrophage (TAM) polarization, nPGSA elicits a potent antigen-specific cellular immune response and sustained tumor regression in melanoma- and neuroblastoma-bearing mice. Our study provides a promising strategy for the delivery of cGAMP, and it offers insights into the function of cGAMP in modulating the tumor immune microenvironment for cancer immunotherapy.
Collapse
Affiliation(s)
- Xiao Lu
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Heming Xia
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences Peking University, Beijing 100191, China
| | - Wei Gao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas 77204-5037, United States
| | - Xingquan Pan
- College of New Materials and Chemical Engineering, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Yue Yan
- Department of Central Laboratory, Peking University First Hospital, Beijing 100034, China
| | - Ran Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yubin He
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences Peking University, Beijing 100191, China
| | - Binlong Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems School of Pharmaceutical Sciences Peking University, Beijing 100191, China
| | - Dong Mei
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| |
Collapse
|
17
|
Prakash G, Parmar B, Bhatia D. Structurally programmable, functionally tuneable dendrimers in biomedical applications. Biomater Sci 2025; 13:875-895. [PMID: 39804192 DOI: 10.1039/d4bm01475h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
The application of nanotechnology in medical biology has seen a significant rise in recent years because of the introduction of novel tools that include supramolecular systems, complexes, and composites. Dendrimers are one of the remarkable examples of such tools. These spherical, regularly branching structures with enhanced cell compatibility and bioavailability have shown to be an excellent option for gene or drug administration. They are the fourth important architectural group of polymers after the three well-known types (branched, cross-linked, and linear polymers). These tiny macromolecules generate nanometer-size structures consisting of branching, with identical units assembled around a central core. By regulating dendrimer synthesis, it is possible to manipulate both their molecular weight and chemical content carefully, permitting predictable tailoring of their biocompatibility and pharmacokinetics, making them a promising candidate for biomedical uses. In contrast to their more easily obtainable synthetic techniques and comparable functions in hyperbranched polymers, dendrimers have demonstrated efficacy in biological applications, exhibiting remarkable sample purity and synthesizing reproducibility. Dendrimers are appealing as basic materials for manufacturing nanomaterials for uses in many different disciplines because of their highly specified chemical structure and globular form. Thus, much effort has been made to create functional materials with dendrimers. Especially looking at dendrimer-based nanomaterials meant for use in the biomedical domain, this review discusses the design, types, properties, and function of bionanomaterials employing several techniques, including surface modification, assembly, and hybrid development, and their uses.
Collapse
Affiliation(s)
- Geethu Prakash
- Department of Biological Sciences and Engineering Indian Institute of Technology, Palaj, Gandhinagar 382355, India.
| | - Bhagyesh Parmar
- Department of Biological Sciences and Engineering Indian Institute of Technology, Palaj, Gandhinagar 382355, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering Indian Institute of Technology, Palaj, Gandhinagar 382355, India.
| |
Collapse
|
18
|
Palmer CR, Pastora LE, Kimmel BR, Pagendarm HM, Kwiatkowski AJ, Stone PT, Arora K, Francini N, Fedorova O, Pyle AM, Wilson JT. Covalent Polymer-RNA Conjugates for Potent Activation of the RIG-I Pathway. Adv Healthc Mater 2025; 14:e2303815. [PMID: 38648653 PMCID: PMC11493851 DOI: 10.1002/adhm.202303815] [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: 11/02/2023] [Revised: 04/13/2024] [Indexed: 04/25/2024]
Abstract
RNA ligands of retinoic acid-inducible gene I (RIG-I) are a promising class of oligonucleotide therapeutics with broad potential as antiviral agents, vaccine adjuvants, and cancer immunotherapies. However, their translation has been limited by major drug delivery barriers, including poor cellular uptake, nuclease degradation, and an inability to access the cytosol where RIG-I is localized. Here this challenge is addressed by engineering nanoparticles that harness covalent conjugation of 5'-triphospate RNA (3pRNA) to endosome-destabilizing polymers. Compared to 3pRNA loaded into analogous nanoparticles via electrostatic interactions, it is found that covalent conjugation of 3pRNA improves loading efficiency, enhances immunostimulatory activity, protects against nuclease degradation, and improves serum stability. Additionally, it is found that 3pRNA could be conjugated via either a disulfide or thioether linkage, but that the latter is only permissible if conjugated distal to the 5'-triphosphate group. Finally, administration of 3pRNA-polymer conjugates to mice significantly increases type-I interferon levels relative to analogous carriers that use electrostatic 3pRNA loading. Collectively, these studies have yielded a next-generation polymeric carrier for in vivo delivery of 3pRNA, while also elucidating new chemical design principles for covalent conjugation of 3pRNA with potential to inform the further development of therapeutics and delivery technologies for pharmacological activation of RIG-I.
Collapse
Affiliation(s)
- Christian R. Palmer
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Lucinda E. Pastora
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Blaise R. Kimmel
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Hayden M. Pagendarm
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | | | - Payton T. Stone
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Karan Arora
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Nora Francini
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
| | - Olga Fedorova
- Department of MolecularCellular and Developmental BiologyYale UniversityNew HavenCT06511USA
- Howard Hughes Medical InstituteChevy ChaseMD20815USA
| | - Anna M. Pyle
- Department of MolecularCellular and Developmental BiologyYale UniversityNew HavenCT06511USA
- Howard Hughes Medical InstituteChevy ChaseMD20815USA
- Department of ChemistryYale UniversityNew HavenCT06511USA
| | - John T. Wilson
- Department of Chemical and Biomolecular EngineeringVanderbilt UniversityNashvilleTN37235USA
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTN37235USA
- Department of PathologyMicrobiologyand ImmunologyVanderbilt University Medical CenterNashvilleTN37232USA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTN37232USA
| |
Collapse
|
19
|
Fan J, Xiao Z, Dong Y, Ye F, Qiu Y, Zhang C, Yin X, Li Y, Wang T. Nanocarrier-Mediated RNA Delivery Platform as a Frontier Strategy for Hepatic Disease Treatment: Challenges and Opportunities. Adv Healthc Mater 2025; 14:e2402933. [PMID: 39723654 DOI: 10.1002/adhm.202402933] [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: 08/07/2024] [Revised: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Hepatic diseases cause serious public health problems worldwide, and there is an urgent need to develop effective therapeutic agents. In recent years, significant progress is made in RNA therapy, and RNA molecules, such as mRNAs, siRNAs, miRNAs, and RNA aptamers, are shown to provide significant advantages in the treatment of hepatic diseases. However, the drawbacks of RNAs, such as their poor biological stability, easy degradation by nucleases in vivo, low bioavailability, and low concentrations in target tissues, significantly limit the clinical application of RNA-based drugs. Therefore, exploring and developing effective nanoscale delivery platforms for RNA therapeutics are of immense value. This review focuses on the different types of hepatic diseases and RNA therapeutics, summarizing various nanoscale delivery platforms and their strengths and weaknesses. Finally, the current status and future prospects of nanoscale delivery systems for RNA therapy are discussed.
Collapse
Affiliation(s)
- Jinhui Fan
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Zhicheng Xiao
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Yafen Dong
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, 201200, China
| | - Fei Ye
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Yan Qiu
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, 201200, China
| | - Chuan Zhang
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Xiaolan Yin
- Cancer center, Shanghai 411 hospital, China RongTong Medical Healthcare Group Co. Ltd./411 Hospital, Shanghai University, Shanghai, 200081, China
| | - Yi Li
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| | - Tingfang Wang
- School of Medicine, 411 Hospital of Shanghai University, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
20
|
Allisha J, Das J, Dunnigan T, Sharfstein ST, Datta P. Stipulations of cell and gene therapy and the ties to biomanufacturing. Biotechnol Prog 2025:e3521. [PMID: 39846483 DOI: 10.1002/btpr.3521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/24/2025]
Abstract
Cell and gene therapy (CGT) products are emerging and innovative biopharmaceuticals that hold promise for treating diseases that are otherwise beyond the scope of conventional medicines. The evolution of CGT from a research idea to a promising therapeutic product is due to the complementary advancements across various scientific disciplines. First, the innovations and advancements in gene editing and delivery technology have provided fundamental tools to manipulate genes and cells for therapeutic pursuits. Second, advancements in applied and translational research, including how clinical trials are designed, performed, evaluated, and analyzed, have transformed the technology into a potential therapeutic product. Third, advancements in scaling up the production of CGT products have been critical in delivering the product for preclinical studies, clinical trials, and approved treatments. In parallel, regulatory requirements have continuously evolved, with lessons learned from translational studies and biomanufacturing. These combined efforts have transformed CGT products from a promising concept into a reality with the potential to treat a wide range of diseases. However, continued R&D and regulatory oversight are crucial to further improve the safety, efficacy, and accessibility of CGT products.
Collapse
Affiliation(s)
- Justin Allisha
- Department of Life Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | - Juthika Das
- Department of Life Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | - Thomas Dunnigan
- Department of Life Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | - Susan T Sharfstein
- Department of Nanoscale Science and Engineering and The RNA Institute, University at Albany, State University of New York, Albany, New York, USA
| | - Payel Datta
- Department of Life Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| |
Collapse
|
21
|
Sprengel C, David C, Berning L, Nollmann C, Lenz T, Stühler K, Stork B, Heinzel T. Lysosomal activity in response to the incubation of pristine and functionalized carbon nanodots. iScience 2025; 28:111654. [PMID: 39886472 PMCID: PMC11780158 DOI: 10.1016/j.isci.2024.111654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/18/2024] [Accepted: 11/26/2024] [Indexed: 02/01/2025] Open
Abstract
We present functional studies of lysosomes in human cells after uptake of carbon nanodots (CNDs). Even under high CND concentrations, the lysosomal functionality, as characterized via cathepsins B and L as well as the autophagic markers SQSTM1/p62 and LC3B-II, is maintained. Furthermore, branched polyethylenimine (bPEI) molecules have been coupled to the CNDs as a model functionalization or example of a drug. We observe that the bPEI-CND conjugates accumulate to a higher degree in the lysosomes as compared to bPEI or CND alone. Here, changes in the lysosomal size and function are observed, which can be explained exclusively by the bPEI. It is concluded that CNDs are highly efficient and inert carriers for functional molecules into lysosomes as target, with the added value that lysosomal escape is suppressed, thereby avoiding unintended side effects in other cellular compartments.
Collapse
Affiliation(s)
- Carla Sprengel
- Solid State Physics Laboratory, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| | - Céline David
- Institute of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Lena Berning
- Institute of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Cathrin Nollmann
- Solid State Physics Laboratory, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| | - Thomas Lenz
- Molecular Proteomics Laboratory, Biological Medical Research Center, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Biological Medical Research Center, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Institute of Molecular Medicine I, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Björn Stork
- Institute of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Thomas Heinzel
- Solid State Physics Laboratory, Heinrich Heine University Düsseldorf, 40204 Düsseldorf, Germany
| |
Collapse
|
22
|
Zheng D, Chen T, Yang K, Yin G, Chen Y, Gui J, Xu C, Lv S. Microfluidic Synthesis of miR-200c-3p Lipid Nanoparticles: Targeting ZEB2 to Alleviate Chondrocyte Damage in Osteoarthritis. Int J Nanomedicine 2025; 20:505-521. [PMID: 39830158 PMCID: PMC11742371 DOI: 10.2147/ijn.s491711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Osteoarthritis (OA) is a degenerative joint disease characterized by articular cartilage degeneration. Chondrocyte inflammation, apoptosis, and extracellular matrix degradation accelerated OA progression. MicroRNA (miRNA) has the potential to be a therapeutic method for osteoarthritis. However, it is difficult to penetrate the cell to exercise its biological function, and its extracellular effect is unclear. Methods lipo-AgPEI-miR-200c-3p was created by combining miR-200c-3p with silver nitrate polyvinylimine nanoparticles on a microfluidic device. The drug release curve, stability, temperature sensitivity, cytotoxicity, and the impact of lipo-AgPEI-miR-200c-3p on the expression of proteins linked to matrix disintegration, apoptosis, and inflammatory factors were all detected. Results Results showed that the particle size of Lipo-AgPEI-miR-200c-3p was about 130 nm, the Zeta potential was lowered to 1.08±0.12 mV. Lipo-AgPEI-miR-200c-3p could increase cell viability, prevent cell apoptosis, and decrease the expression levels of TNF-α, IL-6, IL-1β, and MCP-1 in ADTC5 cells following LPS stimulation. MMP3, MMP13, and ADAMTS-4 expression was downregulated whereas collagen II expression was upregulated. The ZEB2 expression was greatly elevated following LPS stimulation and dramatically decreased following transfection of miR-200c-3p. Collagen II expression rose following transfection of si-ZEB2, whereas the expression levels of inflammatory factors, apoptosis-related proteins, MMP3, MMP13, and ADAMTS-4 decreased. The dual luciferase experiment demonstrated that ZEB2 was the target gene of miR-200c-3p. Conclusion The synergistic effect of AgPEI and miR-200c-3p can inhibit the inflammatory response, apoptosis, and matrix degradation of chondrocytes. Lipo-AgPEI-miR-200c-3p can also improve transfection efficiency and obtain good physicochemical properties of drugs. miR-200c-3p may be crucial in the development of OA and can influence the target gene ZEB2, control the inflammatory response, apoptosis, and chondrocyte matrix breakdown.
Collapse
Affiliation(s)
- Dong Zheng
- Department of Orthopedics, The Affiliated Changzhou No.2 People’s Hospital with Nanjing Medical University, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Tong Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Kaiyuan Yang
- Department of Orthopedics, The Affiliated Changzhou No.2 People’s Hospital with Nanjing Medical University, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Guangrong Yin
- Department of Orthopedics, The Affiliated Changzhou No.2 People’s Hospital with Nanjing Medical University, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Yang Chen
- Changzhou Productivity Development Center, Changzhou, People’s Republic of China
| | - Jianchao Gui
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Chao Xu
- Department of Orthopedics, The Affiliated Changzhou No.2 People’s Hospital with Nanjing Medical University, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Songwei Lv
- School of Pharmacy, Changzhou University, Changzhou, People’s Republic of China
| |
Collapse
|
23
|
Shariati A, Khezrpour A, Shariati F, Afkhami H, Yarahmadi A, Alavimanesh S, Kamrani S, Modarressi MH, Khani P. DNA vaccines as promising immuno-therapeutics against cancer: a new insight. Front Immunol 2025; 15:1498431. [PMID: 39872522 PMCID: PMC11769820 DOI: 10.3389/fimmu.2024.1498431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Cancer is one of the leading causes of mortality around the world and most of our conventional treatments are not efficient enough to combat this deadly disease. Harnessing the power of the immune system to target cancer cells is one of the most appealing methods for cancer therapy. Nucleotide-based cancer vaccines, especially deoxyribonucleic acid (DNA) cancer vaccines are viable novel cancer treatments that have recently garnered significant attention. DNA cancer vaccines are made of plasmid molecules that encode tumor-associated or tumor-specific antigens (TAAs or TSAs), and possibly some other immunomodulatory adjuvants such as pro-inflammatory interleukins. Following the internalization of plasmids into cells, their genes are expressed and the tumor antigens are loaded on major histocompatibility molecules to be presented to T-cells. After the T-cells have been activated, they will look for tumor antigens and destroy the tumor cells upon encountering them. As with any other treatment, there are pros and cons associated with using these vaccines. They are relatively safe, usually well-tolerated, stable, easily mass-produced, cost-effective, and easily stored and transported. They can induce a systemic immune response effective on both the primary tumor and metastases. The main disadvantage of DNA vaccines is their poor immunogenicity. Several approaches including structural modification, combination therapy with conventional and novel cancer treatments (such as chemotherapy, radiotherapy, and immune checkpoint blockade (ICB)), and the incorporation of adjuvants into the plasmid structure have been studied to enhance the vaccine's immunogenicity and improve the clinical outcome of cancer patients. In this review, we will discuss some of the most promising optimization strategies and examine some of the important trials regarding these vaccines.
Collapse
Affiliation(s)
- Alireza Shariati
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Arya Khezrpour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Fatemeh Shariati
- Department of Genetics, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
24
|
Uddin MN, Dinar MAM, Schrass LE, Pack DW, DeRouchey JE. Impact of Acetylation, Succinylation, and pH on DNA Packaging in PEI-Based Polyplexes. Biomacromolecules 2025; 26:178-189. [PMID: 39690700 DOI: 10.1021/acs.biomac.4c00648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Polyethylenimine (PEI) is a widely used cationic polymer for nonviral gene delivery, often modified to enhance transfection efficiency and reduce cytotoxicity. This study investigates how acetylation, succinylation (acPEI and zPEI), and pH influence the internal DNA packaging of polyplexes. Both modifications alter physicochemical properties, leading to complexes that decondense more readily with increasing modification. X-ray scattering reveals that high acetylation produces loosely packed DNA, while succinylation unexpectedly tightens DNA packing at higher modification levels. Polyplexes formed at low pH (pH 4) are more stable and tightly packed than those formed at pH 7.5. Acidifying polyplexes initially formed at pH 7.5 induces structural rearrangement to tighter DNA packing accompanied by significant PEI release, providing direct evidence for models where free PEI aids endosomal escape. These findings challenge conventional assumptions about PEI behavior and offer new insights into DNA packaging, emphasizing tailored polymer modifications and pH conditions to optimize gene delivery.
Collapse
Affiliation(s)
- Md Nasir Uddin
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Md Abu Monsur Dinar
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Leah E Schrass
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Daniel W Pack
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Jason E DeRouchey
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| |
Collapse
|
25
|
Shaulli X, Moreno‐Echeverri AM, Andoni M, Waeber E, Ramakrishna SN, Fritsch C, Vanhecke D, Rothen‐Rutishauser B, Scheffold F. Polymer Nano-Carrier-Mediated Gene Delivery: Visualizing and Quantifying DNA Encapsulation Using dSTORM. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405929. [PMID: 39551983 PMCID: PMC11707562 DOI: 10.1002/smll.202405929] [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: 07/15/2024] [Revised: 09/28/2024] [Indexed: 11/19/2024]
Abstract
The success of gene therapy hinges on the effective encapsulation, protection, and compression of genes. These processes deliver therapeutic genes into designated cells for genetic repair, cellular behavior modification, or therapeutic effect induction. However, quantifying the encapsulation efficiency of small molecules of interest like DNA or RNA into delivery carriers remains challenging. This work shows how super-resolution microscopy, specifically direct stochastic optical reconstruction microscopy (dSTORM), can be employed to visualize and measure the quantity of DNA entering a single carrier. Utilizing pNIPAM/bPEI microgels as model nano-carriers to form polyplexes, DNA entry into the carrier is revealed across different charge ratios at temperatures below and above the volume phase transition of the microgel core. The encapsulation efficiency also depends on DNA length and shape. This work demonstrates the uptake of the carrier entity by primary derived macro-phages and showcases the cell viability of the polyplexes. The study shows that dSTORM is a potent tool for fine-tuning and creating polyplex microgel carrier systems with precise size, shape, and loading capacity at the individual particle level. This advancement shall contribute significantly to optimizing gene delivery systems.
Collapse
Affiliation(s)
- Xhorxhina Shaulli
- Department of PhysicsUniversity of FribourgChemin du Musée 3FribourgCH 1700Switzerland
| | | | - Mariza Andoni
- Department of PhysicsUniversity of FribourgChemin du Musée 3FribourgCH 1700Switzerland
| | - Eileen Waeber
- Department of PhysicsUniversity of FribourgChemin du Musée 3FribourgCH 1700Switzerland
| | | | - Cornelia Fritsch
- Department of BiologyUniversity of FribourgChemin du Musée 10FribourgCH 1700Switzerland
| | - Dimitri Vanhecke
- Adolphe Merkle InstituteUniversity of FribourgChemin des Verdiers 4FribourgCH 1700Switzerland
| | | | - Frank Scheffold
- Department of PhysicsUniversity of FribourgChemin du Musée 3FribourgCH 1700Switzerland
| |
Collapse
|
26
|
Kumari S, Kamiya A, Karnik SS, Rohilla S, Dubey SK, Taliyan R. Novel Gene Therapy Approaches for Targeting Neurodegenerative Disorders: Focusing on Delivering Neurotrophic Genes. Mol Neurobiol 2025; 62:386-411. [PMID: 38856793 DOI: 10.1007/s12035-024-04260-y] [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: 12/06/2023] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
Abstract
Neurodegenerative illnesses (NDDs) like Alzheimer's, Parkinson's, amyotrophic lateral sclerosis, spinal muscular atrophy, and Huntington's disease have demonstrated considerable potential for gene therapy as a viable therapeutic intervention. NDDs are marked by the decline of neurons, resulting in changes in both behavior and pathology within the body. Strikingly, only symptomatic management is available without a cure for the NDDs. There is an unmet need for a permanent therapeutic approach. Many studies have been going on to target the newer therapeutic molecular targets for NDDs including gene-based therapy. Gene therapy has the potential to provide therapeutic benefits to a large number of patients with NDDs by offering mechanisms including neuroprotection, neuro-restoration, and rectification of pathogenic pathways. Gene therapy is a medical approach that aims to modify the biological characteristics of living cells by controlling the expression of specific genes in certain neurological disorders. Despite being the most complex and well-protected organ in the human body, there is clinical evidence to show that it is possible to specifically target the central nervous system (CNS). This provides hope for the prospective application of gene therapy in treating NDDs in the future. There are several advanced techniques available for using viral or non-viral vectors to deliver the therapeutic gene to the afflicted region. Neurotrophic factors (NTF) in the brain are crucial for the development, differentiation, and survival of neurons in the CNS, making them important in the context of various neurological illnesses. Gene delivery of NTF has the potential to be used as a therapeutic approach for the treatment of neurological problems in the brain. This review primarily focuses on the methodologies employed for delivering the genes of different NTFs to treat neurological disorders. These techniques are currently being explored as a viable therapeutic approach for neurodegenerative diseases. The article exclusively addresses gene delivery approaches and does not cover additional therapy strategies for NDDs. Gene therapy offers a promising alternative treatment for NDDs by stimulating neuronal growth instead of solely relying on symptom relief from drugs and their associated adverse effects. It can serve as a long-lasting and advantageous treatment choice for the management of NDDs. The likelihood of developing NDDs increases with age as a result of neuronal degradation in the brain. Gene therapy is an optimal approach for promoting neuronal growth through the introduction of nerve growth factor genes.
Collapse
Affiliation(s)
- Shobha Kumari
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Aayush Kamiya
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sanika Sanjay Karnik
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | - Sumedha Rohilla
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India
| | | | - Rajeev Taliyan
- Indian Council of Medical Research-Senior Research Fellow (ICMR-SRF), Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Pilani, 333031, Rajasthan, India.
| |
Collapse
|
27
|
Zhang Y, Zhang M, Song H, Dai Q, Liu C. Tumor Microenvironment-Responsive Polymer-Based RNA Delivery Systems for Cancer Treatment. SMALL METHODS 2025; 9:e2400278. [PMID: 38803312 DOI: 10.1002/smtd.202400278] [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: 02/25/2024] [Revised: 03/30/2024] [Indexed: 05/29/2024]
Abstract
Ribonucleic acid (RNA) therapeutics offer a broad prospect in cancer treatment. However, their successful application requires overcoming various physiological barriers to effectively deliver RNAs to the target sites. Currently, a number of RNA delivery systems based on polymeric nanoparticles are developed to overcome these barriers in RNA delivery. This work provides an overview of the existing RNA therapeutics for cancer gene therapy, and particularly summarizes those that are entering the clinical phase. This work then discusses the core features and latest research developments of tumor microenvironment-responsive polymer-based RNA delivery carriers which are designed based on the pathological characteristics of the tumor microenvironment. Finally, this work also proposes opportunities for the transformation of RNA therapies into cancer immunotherapy methods in clinical applications.
Collapse
Affiliation(s)
- Yahan Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing, 102206, China
| | - Haiqin Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Qiong Dai
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
28
|
Wang H, Cheng Y. Polymers for mRNA Delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70002. [PMID: 39763235 DOI: 10.1002/wnan.70002] [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: 05/30/2024] [Revised: 10/22/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
mRNA delivery has emerged as a transformative approach in biotechnology and medicine, offering a versatile platform for the development of novel therapeutics. Unlike traditional small molecule drugs or protein-based biologics, mRNA therapeutics have the unique ability to direct cells to generate therapeutic proteins, allowing for precise modulation of biological processes. The delivery of mRNA into target cells is a critical step in realizing the therapeutic potential of this technology. In this review, our focus is on the latest advancements in designing functional polymers to achieve efficient mRNA delivery. Biodegradable polymers and low molecular weight polymers in addressing the balance in mRNA binding and release are summarized. Benefiting from the excellent performance of lipid nanoparticles in mRNA delivery, polymer/lipid hybrid nanostructures are also included. Finally, the challenges and future prospects in the development of polymer-based mRNA delivery systems are discussed.
Collapse
Affiliation(s)
- Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, South China University of Technology, Guangzhou, China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
29
|
Sanati M, Figueroa-Espada CG, Han EL, Mitchell MJ, Yavari SA. Bioengineered Nanomaterials for siRNA Therapy of Chemoresistant Cancers. ACS NANO 2024; 18:34425-34463. [PMID: 39666006 DOI: 10.1021/acsnano.4c11259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Chemoresistance remains a long-standing challenge after cancer treatment. Over the last two decades, RNA interference (RNAi) has emerged as a gene therapy modality to sensitize cancer cells to chemotherapy. However, the use of RNAi, specifically small-interfering RNA (siRNA), is hindered by biological barriers that limit its intracellular delivery. Nanoparticles can overcome these barriers by protecting siRNA in physiological environments and facilitating its delivery to cancer cells. In this review, we discuss the development of nanomaterials for siRNA delivery in cancer therapy, current challenges, and future perspectives for their implementation to overcome cancer chemoresistance.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 97178, Iran
| | - Christian G Figueroa-Espada
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Emily L Han
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| |
Collapse
|
30
|
de Pinho SS, Invenção MDCV, Silva AJD, de Macêdo LS, Espinoza BCF, Leal LRS, da Gama MATM, de Moura IA, Silva MEDS, de Souza DVS, Lara ML, Alves JNSA, de Freitas AC. Pichia pastoris-Derived β-Glucan Capsules as a Delivery System for DNA Vaccines. Vaccines (Basel) 2024; 12:1428. [PMID: 39772088 PMCID: PMC11728682 DOI: 10.3390/vaccines12121428] [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: 10/31/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES DNA vaccines are rapidly produced and adaptable to different pathogens, but they face considerable challenges regarding stability and delivery to the cellular target. Thus, effective delivery methods are essential for the success of these vaccines. Here, we evaluated the efficacy of capsules derived from the cell wall of the yeast Pichia pastoris as a delivery system for DNA vaccines. METHODS The capsules were extracted from the yeast Pichia pastoris strain GS115, previously grown in a YPD medium. pVAX1 expression vector was adopted to evaluate the DNA vaccine insertion and delivery. Three encapsulation protocols were tested to identify the most effective in internalizing the plasmid. The presence of plasmids inside the capsules was confirmed by fluorescence microscopy, and the encapsulation efficiency was calculated by the difference between the initial concentration of DNA used for insertion and the concentration of unencapsulated DNA contained in the supernatant. The capsules were subjected to different temperatures to evaluate their thermostability and were co-cultured with macrophages for phagocytosis analysis. HEK-293T cells were adopted to assess the cytotoxicity levels by MTT assay. RESULTS The microscopy results indicated that the macrophages successfully phagocytosed the capsules. Among the protocols tested for encapsulation, the one with 2% polyethylenimine for internalization showed the highest efficiency, with an encapsulation rate above 80%. However, the vaccine capsules obtained with the protocol that used 5% NaCl showed better thermal stability and encapsulation efficiency above 63% without induction of cell viability loss in HEK 293T. CONCLUSIONS We successfully described a vaccine delivery system using yeast capsules derived from Pichia pastoris, demonstrating its potential for DNA vaccine delivery for the first time. Additional studies will be needed to characterize and improve this delivery strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (S.S.d.P.); (M.d.C.V.I.); (A.J.D.S.); (L.S.d.M.); (B.C.F.E.); (L.R.S.L.); (M.A.T.M.d.G.); (I.A.d.M.); (M.E.d.S.S.); (D.V.S.d.S.); (M.L.L.); (J.N.S.A.A.)
| |
Collapse
|
31
|
Bayraktutan H, Symonds P, Brentville VA, Moloney C, Galley C, Bennett CL, Mata A, Durrant L, Alexander C, Gurnani P. Sparsely PEGylated poly(beta-amino ester) polyplexes enhance antigen specific T-cell response of a bivalent SARS-CoV-2 DNA vaccine. Biomaterials 2024; 311:122647. [PMID: 38878479 DOI: 10.1016/j.biomaterials.2024.122647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 08/06/2024]
Abstract
DNA technology has emerged as a promising route to accelerated manufacture of sequence agnostic vaccines. For activity, DNA vaccines must be protected and delivered to the correct antigen presenting cells. However, the physicochemical properties of the vector must be carefully tuned to enhance interaction with immune cells and generate sufficient immune response for disease protection. In this study, we have engineered a range of polymer-based nanocarriers based on the poly(beta-amino ester) (PBAE) polycation platform to investigate the role that surface poly(ethylene glycol) (PEG) density has on pDNA encapsulation, formulation properties and gene transfectability both in vitro and in vivo. We achieved this by synthesising a non-PEGylated and PEGylated PBAE and produced formulations containing these PBAEs, and mixed polyplexes to tune surface PEG density. All polymers and co-formulations produced small polyplex nanoparticles with almost complete encapsulation of the cargo in all cases. Despite high gene transfection in HEK293T cells, only the fully PEGylated and mixed formulations displayed significantly higher expression of the reporter gene than the negative control in dendritic cells. Further in vivo studies with a bivalent SARS-CoV-2 pDNA vaccine revealed that only the mixed formulation led to strong antigen specific T-cell responses, however this did not translate into the presence of serum antibodies indicating the need for further studies into improving immunisation with polymer delivery systems.
Collapse
Affiliation(s)
- Hulya Bayraktutan
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK; Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Peter Symonds
- Scancell Ltd, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Victoria A Brentville
- Scancell Ltd, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Cara Moloney
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK; Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Charlotte Galley
- Department of Haematology, UCL Cancer Institute, 72 Huntley Street, University College London, London, WC1E 6DD, UK
| | - Clare L Bennett
- Department of Haematology, UCL Cancer Institute, 72 Huntley Street, University College London, London, WC1E 6DD, UK
| | - Alvaro Mata
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, NG7 2RD, UK; Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Lindy Durrant
- Scancell Ltd, University of Nottingham Biodiscovery Institute, Nottingham, NG7 2RD, UK
| | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK.
| | - Pratik Gurnani
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK.
| |
Collapse
|
32
|
O'Hare M, Miller WP, Arevalo-Alquichire S, Amarnani D, Apryani E, Perez-Corredor P, Marino C, Shu DY, Vanderleest TE, Muriel-Torres A, Gordon HB, Gunawan AL, Kaplan BA, Barake KW, Bejjani RP, Doan TH, Lin R, Delgado-Tirado S, Gonzalez-Buendia L, Rossin EJ, Zhao G, Eliott D, Weinl-Tenbruck C, Chevessier-Tünnesen F, Rejman J, Montrasio F, Kim LA, Arboleda-Velasquez JF. An mRNA-encoded dominant-negative inhibitor of transcription factor RUNX1 suppresses vitreoretinal disease in experimental models. Sci Transl Med 2024; 16:eadh0994. [PMID: 39602510 DOI: 10.1126/scitranslmed.adh0994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/06/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
Messenger RNA (mRNA)-based therapies are a promising approach to medical treatment. Except for infectious diseases, no other disease has mRNA-based therapies available. The eye is an ideal model for mRNA therapeutic development because it requires limited dosing. Proliferative vitreoretinopathy (PVR) is a blinding condition caused by retinal detachment that now lacks available medical treatment, with surgery as the only treatment option. We previously implicated runt-related transcription factor-1 (RUNX1) as a driver of epithelial-to-mesenchymal transition (EMT) in PVR and as a critical mediator of aberrant ocular angiogenesis when up-regulated. On the basis of these findings, an mRNA was designed to express a dominant-negative inhibitor of RUNX1 (RUNX1-Trap). We show that RUNX1-Trap delivered in polymer-lipidoid complexes or lipid nanoparticles sequestered RUNX1 in the cytosol and strongly reduced proliferation in primary cell cultures established from fibrotic membranes derived from patients with PVR. We assessed the preclinical efficacy of intraocular delivery of mRNA-encoded RUNX1-Trap in a rabbit model of PVR and in a laser-induced mouse model of aberrant angiogenesis often used to study wet age-related macular degeneration. mRNA-encoded RUNX1-Trap suppressed ocular pathology, measured as pathological scores in the rabbit PVR model and leakage and lesion size in the laser-induced choroidal neovascularization mouse model. mRNA-encoded RUNX1-Trap also strongly reduced proliferation in a human ex vivo explant model of PVR. These data demonstrate the therapeutic potential of mRNA-encoded therapeutic molecules with dominant-negative properties, highlighting the potential of mRNA-based therapies beyond standard gene supplementation approaches.
Collapse
Affiliation(s)
- Michael O'Hare
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - William P Miller
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Said Arevalo-Alquichire
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Dhanesh Amarnani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Evhy Apryani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Paula Perez-Corredor
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Claudia Marino
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Daisy Y Shu
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Timothy E Vanderleest
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Andres Muriel-Torres
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Harper B Gordon
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Audrey L Gunawan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Bryan A Kaplan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Karim W Barake
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Romy P Bejjani
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Tri H Doan
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Rose Lin
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Santiago Delgado-Tirado
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Lucia Gonzalez-Buendia
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Elizabeth J Rossin
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Guannan Zhao
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Dean Eliott
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | - Leo A Kim
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
- Mass Eye and Ear and the Department of Ophthalmology at Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
33
|
Lu X, Qiu J, Li Y, Cai M, Yang X, Li S, Ye G, Yi W, Huang Y. PEGylation Can Effectively Strike a Balance in siRNA Delivery Performances of Guanidinylated Linear Synthetic Polypeptides with Potential Use for Transcriptional Gene Silencing. ACS Macro Lett 2024; 13:1251-1257. [PMID: 39259674 DOI: 10.1021/acsmacrolett.4c00405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
The prevailing design philosophy for polymeric vectors delivering siRNA is rooted in the post-transcriptional gene silencing (PTGS) mechanism. Yet, the transcriptional gene silencing (TGS) mechanism offers a potentially more durable silencing effect, which necessitates efficient siRNA delivery into the nucleus. However, it remains a challenge for the polymeric vectors to efficiently deliver siRNA into the nucleus. We have explored guanidinylated cyclic synthetic polypeptides (GCSPs) to enhance the nuclear delivery of siRNA, but an increased cytotoxicity and difficulty in producing the GCSPs on a large scale limit their utility. Herein, we simply prepare PEGylated guanidinylated linear synthetic polypeptides (PGLSPs) exhibiting improved membrane penetration, direct siRNA transport to the nucleus, reduced toxicity, high cellular uptake, and mitigation of protein corona formation. The PEGylation can effectively balance the vector's nuclear delivery capacity with other critical aspects of performances for siRNA delivery. Therefore, the PGLSPs hold promise as TGS-based delivery vectors, offering potential for future therapeutic applications.
Collapse
Affiliation(s)
- Xujun Lu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiajian Qiu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yilan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Ming Cai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaohan Yang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Suifei Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Guodong Ye
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Wei Yi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yugang Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
34
|
Qin C, Wang H, Cui H, Wang Y, Zhang M, Li H, Liu Y, Wang J, Chen Q, Zhao Y. Synthetic genomic nanomedicine with triple-responsiveness for systemic anti-tumor therapy. J Colloid Interface Sci 2024; 672:350-362. [PMID: 38850862 DOI: 10.1016/j.jcis.2024.06.010] [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: 02/12/2024] [Revised: 05/26/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
To overcome the biological barriers in the journey of systemic gene delivery, a multifaceted genomic synthetic nanomedicine was elaborated and strategically equipped with a multiple of intriguing responsiveness. Particularly, core-shell plasmid DNA condensates were created based on polyionic complexation with block copolymer of polyethylene glycol (PEG)-polylysine (PLys), namely, the nanoscaled PLys&pDNA nanoparticle tethered with the biocompatible PEG surroundings. Furthermore, redox-reversible disulfide crosslinking was introduced into PLys&pDNA nanoparticle to accomplish adequate structural stabilities, and thermal-responsive polypropylacrylamide (PNIPAM) was introduced as the secondary intermediate surroundings onto the pre-formulated PLys&pDNA nanoparticle with the aim of preventing the potential enzymatic degradation from the environmental nucleases. Hence, hundreds of times prolonged survival and retention was determined in pertinent to the blood circulation properties. Additionally, the installation of a guide ligand at the distal end of PEG segments was proposed to encourage selective tumor uptake. A linear peptide of GPLGVRG, which is selectively susceptible to digestion by the tumor-enriched matrix metalloproteinase 2 (MMP-2), was used as the linkage between the shell and core. This peptide has been shown to detach the bio-inert PEGylation, resulting in further facilitated cell endocytosis and intracellular trafficking activities. Hence, the precisely defined synthetic nanomedicine, which exhibits desirable characteristics, efficient expression of the therapeutic gene in the affected cells, and contributed to potent therapeutic efficacy in systemic treatment of intractable tumors by encapsulating the anti-angiogenic gene.
Collapse
Affiliation(s)
- Chunfang Qin
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Hao Wang
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Hongyan Cui
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China
| | - Yue Wang
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang City, Liaoning 110042, China; Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang City, Liaoning 110042, China; Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang City, Liaoning 110042, China; China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, Liaoning Province 110122, China
| | - Ming Zhang
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang City, Liaoning 110042, China
| | - Haidong Li
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Yuchen Liu
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Jingyun Wang
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China.
| | - Qixian Chen
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang 314100, China; Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang City, Liaoning 110042, China.
| | - Yan Zhao
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang City, Liaoning 110042, China; Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang City, Liaoning 110042, China; Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang City, Liaoning 110042, China; China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, Liaoning Province 110122, China.
| |
Collapse
|
35
|
Padhy A, Das P, Mahadik NS, Panda S, Anas M, Das S, Banerjee R, Sen Gupta S. Design and synthesis of a shikimoyl-functionalized cationic di-block copolypeptide for cancer cell specific gene transfection. J Mater Chem B 2024; 12:8952-8965. [PMID: 39171401 DOI: 10.1039/d4tb01233j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Targeted and efficient gene delivery systems hold tremendous potential for the improvement of cancer therapy by enabling appropriate modification of biological processes. Herein, we report the design and synthesis of a novel cationic di-block copolypeptide, incorporating homoarginine (HAG) and shikimoyl (LSA) functionalities (HDA-b-PHAGm-b-PLSAn), tailored for enhanced gene transfection specifically in cancer cells. The di-block copolypeptide was synthesized via sequential N-carboxyanhydride (NCA) ring-opening polymerization (ROP) techniques and its physicochemical properties were characterized, including molecular weight, dispersity, secondary conformation, size, morphology, and surface charge. In contrast to the cationic poly-L-homoarginine, we observed a significantly reduced cytotoxic effect of this di-block copolypeptide due to the inclusion of the shikimoyl glyco-polypeptide block, which also added selectivity in internalizing particular cells. This di-block copolypeptide was internalized via mannose-receptor-mediated endocytosis, which was investigated by competitive receptor blocking with mannan. We evaluated the transfection efficiency of the copolypeptide in HEK 293T (noncancerous cells), MDA-MB-231 (breast cancer cells), and RAW 264.7 (dendritic cells) and compared it with commonly employed transfection agents (Lipofectamine). Our findings demonstrate that the homoarginine and shikimoyl-functionalized cationic di-block copolypeptide exhibits potent gene transfection capabilities with minimal cytotoxic effects, particularly in cancer cells, while it is ineffective for normal cells, indicative of its potential as a promising platform for cancer cell-specific gene delivery systems. To evaluate this, we delivered an artificially designed miRNA-plasmid against Hsp90 (amiR-Hsp90) which upon successful transfection depleted the Hsp90 (a chaperone protein responsible for tumour growth) level specifically in cancerous cells and enforced apoptosis. This innovative approach offers a new avenue for the development of targeted therapeutics with an improved efficacy and safety profile in cancer treatment.
Collapse
Affiliation(s)
- Abinash Padhy
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741246, India.
| | - Pritam Das
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Namita S Mahadik
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Sidharth Panda
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741246, India.
| | - Mahammad Anas
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700 032, India
| | - Sabyasachi Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741246, India.
| | - Rajkumar Banerjee
- Department of Oils, Lipids Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Sayam Sen Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal 741246, India.
| |
Collapse
|
36
|
Khan M. Polymers as Efficient Non-Viral Gene Delivery Vectors: The Role of the Chemical and Physical Architecture of Macromolecules. Polymers (Basel) 2024; 16:2629. [PMID: 39339093 PMCID: PMC11435517 DOI: 10.3390/polym16182629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Gene therapy is the technique of inserting foreign genetic elements into host cells to achieve a therapeutic effect. Although gene therapy was initially formulated as a potential remedy for specific genetic problems, it currently offers solutions for many diseases with varying inheritance patterns and acquired diseases. There are two major groups of vectors for gene therapy: viral vector gene therapy and non-viral vector gene therapy. This review examines the role of a macromolecule's chemical and physical architecture in non-viral gene delivery, including their design and synthesis. Polymers can boost circulation, improve delivery, and control cargo release through various methods. The prominent examples discussed include poly-L-lysine, polyethyleneimine, comb polymers, brush polymers, and star polymers, as well as hydrogels and natural polymers and their modifications. While significant progress has been made, challenges still exist in gene stabilization, targeting specificity, and cellular uptake. Overcoming cytotoxicity, improving delivery efficiency, and utilizing natural polymers and hybrid systems are vital factors for prospects. This comprehensive review provides an illuminating overview of the field, guiding the way toward innovative non-viral-based gene delivery solutions.
Collapse
Affiliation(s)
- Majad Khan
- Department of Chemistry, King Fahd University of Petroleum & Minerals KFUPM, Dahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management (IRC-HTCM), King Fahd University of Petroleum & Minerals KFUPM, Dahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Refining and Advanced Chemicals (IRC-CRAC), King Fahd University of Petroleum & Minerals (KFUPM), Dhahran 31261, Saudi Arabia
| |
Collapse
|
37
|
Kuhn E, Srinageshwar B, Story DT, Swanson D, Sharma A, Dunbar GL, Rossignol J. Delivery of PAMAM dendrimers and dendriplexes across natural barriers (blood-brain barrier and placental barrier) in healthy pregnant mice. DISCOVER NANO 2024; 19:148. [PMID: 39264474 PMCID: PMC11393257 DOI: 10.1186/s11671-024-04105-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Gene therapy is an important tool for treating fetal diseases that allows for the delivery and integration of therapeutic genes into the genome of cells carrying mutations. Nanomolecules, like PAMAM dendrimers, have recently come into wider use for carrying vectors as they have several advantages over viral vectors. Namely, (1) tunable size and surface chemistry, (2) uniform size, (3) the ability to target specific tissues, and (4) the ability to carry large biomolecules and drugs. Recently, we demonstrated that 4th generation (G4) PAMAM dendrimer with a cystamine core and a non-toxic surface having 90% -OH and 10% -NH2 groups (D-Cys) could cross the blood-brain barrier following injection into the bloodstream. In the current study, as a proof of concept, we delivered the dendrimers alone (D-Cys) tagged with Cy5.5 (D-Cys-cy5.5) to healthy pregnant C57BL/6J mice to determine the fate of these dendrimers in the pregnant mice as well as in the fetus. Systematic diffusion of the D-Cys-cy5.5 was evaluated on gestational day 17 (3 days after injection) using in vivo imaging. This revealed that the dendrimer was taken up into circulation and away from the injection site. Analysis of sections by fluorescence microscopy showed that D-Cys-cy5.5 was able to successfully cross the maternal blood-brain barrier. However, analysis of the fetal brains failed to detect dendrimers in the central nervous system (CNS). Instead, they appeared to be retained in the placenta. This is one of the first studies to analyze the distribution of surface-modified PAMAM dendrimer in the pregnant mouse and fetus following systemic injection.
Collapse
Affiliation(s)
- Eric Kuhn
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Bhairavi Srinageshwar
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Darren T Story
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Department of Psychology, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Department of Psychology, Saginaw Valley State University, University Center, MI, 48710, USA
| | - Douglas Swanson
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Ajit Sharma
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Gary L Dunbar
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
- Department of Psychology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Julien Rossignol
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA.
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, 48859, USA.
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA.
| |
Collapse
|
38
|
Tang Q, Tomás RMF, Gibson MI. Covalent recruitment of polymers and nanoparticles onto glycan-engineered cells enhances gene delivery during short exposure. Chem Sci 2024:d4sc03666b. [PMID: 39263661 PMCID: PMC11382546 DOI: 10.1039/d4sc03666b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
Non-viral gene delivery with cationic polymers/nanoparticles relies on iterative optimization of the carrier to achieve delivery. Here we demonstrate, instead, that precision engineering of cell surfaces to covalently capture a polyplex accelerates gene delivery within just 10 min of exposure. Azides were installed into cell-surface sialic acids, which enabled the rapid and selective recruitment of cyclooctyne-functional polyplexes, leading to increased delivery of fluorescent cargo, and also increased plasmid expression and siRNA knockdown. Covalent delivery enhancement was also shown for a polymer-coated nanoparticle delivery system. This validates using cellular metabolic engineering (or other synthetic biology) tools to overcome payload delivery challenges.
Collapse
Affiliation(s)
- Qiao Tang
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
- Department of Chemistry, University of Manchester Oxford Road Manchester M13 9PL UK
- Cryologyx Ltd 71-75 Shelton Street London WC2H 9JQ UK
| | - Ruben M F Tomás
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
- Cryologyx Ltd 71-75 Shelton Street London WC2H 9JQ UK
| | - Matthew I Gibson
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
- Department of Chemistry, University of Manchester Oxford Road Manchester M13 9PL UK
- Manchester Institute of Biotechnology 131 Princess Street Manchester M1 7DN UK
| |
Collapse
|
39
|
Liu W, Hou B, Ma D, Peng M, Mao H, Liu W, Gu Z, Chen S. Virus mimicking liposomes incorporated microneedles delivery platform for efficient skin penetration and enhanced cellular uptake. J Drug Deliv Sci Technol 2024; 99:105953. [DOI: 10.1016/j.jddst.2024.105953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
40
|
Mapfumo P, Reichel LS, Leer K, Egger J, Dzierza A, Peneva K, Fischer D, Traeger A. Harnessing Guanidinium and Imidazole Functional Groups: A Dual-Charged Polymer Strategy for Enhanced Gene Delivery. ACS Macro Lett 2024; 13:1000-1007. [PMID: 39052525 PMCID: PMC11340021 DOI: 10.1021/acsmacrolett.4c00321] [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: 05/15/2024] [Revised: 06/30/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Histidine and arginine are two amino acids that exhibit beneficial properties for gene delivery. In particular, the imidazole group of histidine facilitates endosomal release, while the guanidinium group of arginine promotes cellular entry. Consequently, a dual-charged copolymer library based on these amino acids was synthesized via reversible addition-fragmentation chain transfer (RAFT) polymerization. The content of the N-acryloyl-l-histidine (His) monomer was systematically increased, while maintaining consistent levels of methyl N-acryloyl-l-argininate hydrochloride (ArgOMe) or N-(4-guanidinobutyl)acrylamide hydrochloride (GBAm). The resulting polymers formed stable, nanosized polyplexes when complexed with nucleic acids. Remarkably, candidates with increased His content exhibited reduced cytotoxicity profiles and enhanced transfection efficiency, particularly retaining this performance level at lower pDNA concentrations. Furthermore, endosomal release studies revealed that increased His content improved endosomal release, while ArgOMe improved cellular entry. These findings underscore the potential of customized dual-charged copolymers and the synergistic effects of His and ArgOMe/GBAm in enhancing gene delivery.
Collapse
Affiliation(s)
- Prosper
P. Mapfumo
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Liên S. Reichel
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Katharina Leer
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Jan Egger
- Division
of Pharmaceutical Technology and Biopharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, 91058 Erlangen, Germany
| | - Andreas Dzierza
- Division
of Pharmaceutical Technology and Biopharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, 91058 Erlangen, Germany
| | - Kalina Peneva
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
- Center
for Energy and Environmental Chemistry Jena (CEEC), Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Dagmar Fischer
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
- Division
of Pharmaceutical Technology and Biopharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Cauerstr. 4, 91058 Erlangen, Germany
- FAU NeW -
Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Anja Traeger
- Institute
of Organic Chemistry and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| |
Collapse
|
41
|
Neill B, Romero AR, Fenton OS. Advances in Nonviral mRNA Delivery Materials and Their Application as Vaccines for Melanoma Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4894-4913. [PMID: 37930174 PMCID: PMC11220486 DOI: 10.1021/acsabm.3c00721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Messenger RNA (mRNA) vaccines are promising platforms for cancer immunotherapy because of their potential to encode for a variety of tumor antigens, high tolerability, and capacity to induce strong antitumor immune responses. However, the clinical translation of mRNA cancer vaccines can be hindered by the inefficient delivery of mRNA in vivo. In this review, we provide an overview of mRNA cancer vaccines by discussing their utility in treating melanoma. Specifically, we begin our review by describing the barriers that can impede mRNA delivery to target cells. We then review native mRNA structure and discuss various modification methods shown to enhance mRNA stability and transfection. Next, we outline the advantages and challenges of three nonviral carrier platforms (lipid nanoparticles, polymeric nanoparticles, and lipopolyplexes) frequently used for mRNA delivery. Last, we summarize preclinical and clinical studies that have investigated nonviral mRNA vaccines for the treatment of melanoma. In writing this review, we aim to highlight innovative nonviral strategies designed to address mRNA delivery challenges while emphasizing the exciting potential of mRNA vaccines as next-generation therapies for the treatment of cancers.
Collapse
Affiliation(s)
- Bevin Neill
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriana Retamales Romero
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Owen S. Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
42
|
Mapfumo P, Reichel LS, André T, Hoeppener S, Rudolph LK, Traeger A. Optimizing Biocompatibility and Gene Delivery with DMAEA and DMAEAm: A Niacin-Derived Copolymer Approach. Biomacromolecules 2024; 25:4749-4761. [PMID: 38963401 PMCID: PMC11323007 DOI: 10.1021/acs.biomac.4c00007] [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: 01/03/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024]
Abstract
Gene therapy is pivotal in nanomedicine, offering a versatile approach to disease treatment. This study aims to achieve an optimal balance between biocompatibility and efficacy, which is a common challenge in the field. A copolymer library is synthesized, incorporating niacin-derived monomers 2-acrylamidoethyl nicotinate (AAEN) or 2-(acryloyloxy)ethyl nicotinate (AEN) with N,N-(dimethylamino)ethyl acrylamide (DMAEAm) or hydrolysis-labile N,N-(dimethylamino)ethyl acrylate (DMAEA). Evaluation of the polymers' cytotoxicity profiles reveals that an increase in AAEN or DMAEA molar ratios correlates with improved biocompatibility. Remarkably, an increase in AAEN in both DMAEA and DMAEAm copolymers demonstrated enhanced transfection efficiencies of plasmid DNA in HEK293T cells. Additionally, the top-performing polymers demonstrate promising gene expression in challenging-to-transfect cells (THP-1 and Jurkat cells) and show no significant effect on modulating immune response induction in ex vivo treated murine monocytes. Overall, the best performing candidates exhibit an optimal balance between biocompatibility and efficacy, showcasing potential advancements in gene therapy.
Collapse
Affiliation(s)
- Prosper
P. Mapfumo
- Institute
of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, Jena 07743, Germany
| | - Liên S. Reichel
- Institute
of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, Jena 07743, Germany
| | - Thomas André
- Leibniz
Institute on Aging-Fritz Lipmann Institute, Jena 07745, Germany
| | - Stephanie Hoeppener
- Institute
of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, Jena 07743, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, Jena 07743, Germany
| | | | - Anja Traeger
- Institute
of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, Jena 07743, Germany
- Jena
Center for Soft Matter (JCSM), Friedrich
Schiller University Jena, Philosophenweg 7, Jena 07743, Germany
| |
Collapse
|
43
|
Steffens RC, Thalmayr S, Weidinger E, Seidl J, Folda P, Höhn M, Wagner E. Modulating efficacy and cytotoxicity of lipoamino fatty acid nucleic acid carriers using disulfide or hydrophobic spacers. NANOSCALE 2024; 16:13988-14005. [PMID: 38984864 DOI: 10.1039/d4nr01357c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Double pH-responsive xenopeptides comprising polar ionizable succinoyl tetraethylene pentamine (Stp) motifs and lipophilic ionizable lipoamino fatty acids (LAFs) were recently found to efficiently transfect mRNA and pDNA at low doses. However, potency was often accompanied with cytotoxicity at higher doses. Insertion of bioreducible disulfide building blocks (ssbb) or non-reducible hydrophobic spacers between polar and apolar ionizable domains of LAF-Stp carriers should mitigate toxicity of xenopeptides. Carriers showed stable nucleic acid complexation and endosomal pH-dependent lytic activities, both of which were abolished after reductive cleavage of ssbb-containing carriers. For pDNA, U-shaped carriers with one Stp and two LAF units or bundle carriers with two Stps and four LAFs displayed highest potency. For mRNA, best transfection was achieved with bundle carriers with one Stp and four LAFs. Both the ssbb and hydrophobic spacer containing analogs displayed improved metabolic activity, reduced membrane damage, and improved cell growth. The ssbb carriers were most beneficial regarding living cell count and low apoptosis rates. Mechanistically, inserted spacers decelerated the transfection kinetics and altered the requirement of endosomal protonation. Overall, mRNA and pDNA carriers with improved biocompatibility have been designed, with their high potency illustrated in transfection of various cell lines including low passage number colon carcinoma cells.
Collapse
Affiliation(s)
- Ricarda C Steffens
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Sophie Thalmayr
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
- CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany
| | - Eric Weidinger
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
| | - Johanna Seidl
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
- CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany
| | - Paul Folda
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, LMU Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
- CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany
| |
Collapse
|
44
|
Yan X, Chen Q. Polyamidoamine Dendrimers: Brain-Targeted Drug Delivery Systems in Glioma Therapy. Polymers (Basel) 2024; 16:2022. [PMID: 39065339 PMCID: PMC11280609 DOI: 10.3390/polym16142022] [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: 05/23/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Glioma is the most common primary intracranial tumor, which is formed by the malignant transformation of glial cells in the brain and spinal cord. It has the characteristics of high incidence, high recurrence rate, high mortality and low cure rate. The treatments for glioma include surgical removal, chemotherapy and radiotherapy. Due to the obstruction of the biological barrier of brain tissue, it is difficult to achieve the desired therapeutic effects. To address the limitations imposed by the brain's natural barriers and enhance the treatment efficacy, researchers have effectively used brain-targeted drug delivery systems (DDSs) in glioma therapy. Polyamidoamine (PAMAM) dendrimers, as branched macromolecular architectures, represent promising candidates for studies in glioma therapy. This review focuses on PAMAM-based DDSs in the treatment of glioma, highlighting their physicochemical characteristics, structural properties as well as an overview of the toxicity and safety profiles.
Collapse
Affiliation(s)
- Xinyi Yan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Qi Chen
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
45
|
Ge Y, Li W, Tian J, Yu H, Wang Z, Wang M, Dong Z. Single-Stranded Nucleic Acid Transmembrane Molecular Carriers Based on Positively Charged Helical Foldamers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400678. [PMID: 38757406 PMCID: PMC11267351 DOI: 10.1002/advs.202400678] [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: 01/22/2024] [Revised: 04/22/2024] [Indexed: 05/18/2024]
Abstract
Transmembrane delivery of biologically active nucleic acids is an important process in cells and has inspired one to develop advanced drug delivery techniques. In this contribution, molecular-level single-stranded nucleic acid transmembrane carriers are reported based on 3.2 nm long Huc's foldamers (AOrnQ3Q3)8 and (mQ3Q2)8 with linearly and helically aligned positive charges, respectively. These two foldamers not only show very strong DNA affinity via electrostatic interactions but also discriminatively bind single-stranded DNA (ss-DNA) and double-stranded DNA (ds-DNA), corroborating the importance of precise charge arrangement in the electrostatic interactions. More importantly, these two foldamers are capable of efficiently transporting ss-DNA across the lipid membranes, and the ss-DNA transport activity of (AOrnQ3Q3)8 with linearly aligned charges is higher than that of (mQ3Q2)8 with helically aligned charges. Thus a type of novel single-stranded nucleic acid transmembrane molecular carriers based on positively charged helical foldamers are introduced. Further, effective and enhanced expression in EGFP-mRNA transfection experiments strongly demonstrates the potential of positively charged foldamers for RNA transmembrane transport and therapy.
Collapse
Affiliation(s)
- Yunpeng Ge
- State Key Laboratory of Supramolecular Structure and MaterialsCollege of ChemistryJilin University2699 Qianjin StreetChangchun130012P. R. China
- Center for Supramolecular Chemical BiologyJilin University2699 Qianjin StreetChangchun130012P. R. China
| | - Wencan Li
- State Key Laboratory of Supramolecular Structure and MaterialsCollege of ChemistryJilin University2699 Qianjin StreetChangchun130012P. R. China
- Center for Supramolecular Chemical BiologyJilin University2699 Qianjin StreetChangchun130012P. R. China
| | - Jun Tian
- State Key Laboratory of Supramolecular Structure and MaterialsCollege of ChemistryJilin University2699 Qianjin StreetChangchun130012P. R. China
- Center for Supramolecular Chemical BiologyJilin University2699 Qianjin StreetChangchun130012P. R. China
| | - Hao Yu
- State Key Laboratory of Supramolecular Structure and MaterialsCollege of ChemistryJilin University2699 Qianjin StreetChangchun130012P. R. China
| | - Zhenzhu Wang
- State Key Laboratory of Supramolecular Structure and MaterialsCollege of ChemistryJilin University2699 Qianjin StreetChangchun130012P. R. China
- Center for Supramolecular Chemical BiologyJilin University2699 Qianjin StreetChangchun130012P. R. China
| | - Ming Wang
- State Key Laboratory of Supramolecular Structure and MaterialsCollege of ChemistryJilin University2699 Qianjin StreetChangchun130012P. R. China
| | - Zeyuan Dong
- State Key Laboratory of Supramolecular Structure and MaterialsCollege of ChemistryJilin University2699 Qianjin StreetChangchun130012P. R. China
- Center for Supramolecular Chemical BiologyJilin University2699 Qianjin StreetChangchun130012P. R. China
| |
Collapse
|
46
|
Liu J, Chen L, Sun Z, Tao Z, Pavel V, Li Y, Wang F, Cui W, Liu S. Unidirectional gene delivery electrospun fibrous membrane via charge repulsion for tendon repair. Bioact Mater 2024; 37:191-205. [PMID: 38549775 PMCID: PMC10972767 DOI: 10.1016/j.bioactmat.2024.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 11/12/2024] Open
Abstract
Gene therapy is capable of efficiently regulating the expression of abnormal genes in diseased tissues and expected to be a therapeutic option for refractory diseases. However, unidirectional targeting gene therapy is always desired at the tissue interface. In this study, inspired by the principle that like charges repulse each other, a positively charged micro-nano electrospun fibrous membrane with dual-layer structure was developed by electrospinning technology to achieve unidirectional delivery of siRNA-loaded cationic nanocarriers, thus realizing unidirectional gene therapy at the tendon-paratenon interface. Under the charge repulsion of positively charged layer, more cationic COX-2 siRNA nanocarriers were enriched in peritendinous tissue, which not only improved the bioavailability of the gene drug to prevent the peritendinous adhesion formation, but also avoided adverse effects on the fragile endogenous healing of tendon itself. In summary, this study provides an innovative strategy for unidirectional targeting gene therapy of tissue interface diseases by utilizing charge repulsion to facilitate unidirectional delivery of gene drugs.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Liang Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Zhenyu Sun
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Zaijin Tao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| | - Volotovski Pavel
- Republican Scientific and Practical Center of Traumatology and Orthopedics, Belarusian State Medical University, Minsk 220024, Belarus
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Fei Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Shen Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, PR China
| |
Collapse
|
47
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
48
|
Tiwade PB, Ma Y, VanKeulen-Miller R, Fenton OS. A Lung-Expressing mRNA Delivery Platform with Tunable Activity in Hypoxic Environments. J Am Chem Soc 2024; 146:17365-17376. [PMID: 38874565 DOI: 10.1021/jacs.4c04565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Messenger RNA (mRNA) delivery platforms often facilitate protein expression in the liver following intravenous injection and have been optimized for use in normally oxygenated cells (21% O2 atmosphere). However, there is a growing need for mRNA therapy in diseases affecting non-liver organs, such as the lungs. Additionally, many diseases are characterized by hypoxia (<21% O2 atmosphere), a state of abnormally low oxygenation in cells and tissues that can reduce the efficacy of mRNA therapies by upwards of 80%. Here, we report a Tunable Lung-Expressing Nanoparticle Platform (TULEP) for mRNA delivery, whose properties can be readily tuned for optimal expression in hypoxic environments. Briefly, our study begins with the synthesis and characterization of a novel amino acrylate polymer that can be effectively complexed with mRNA payloads into TULEPs. We study the efficacy and mechanism of mRNA delivery using TULEP, including analysis of the cellular association, endocytosis mechanisms, endosomal escape, and protein expression in a lung cell line. We then evaluate TULEP under hypoxic conditions and address hypoxia-related deficits in efficacy by making our system tunable with adenosine triphosphate (ATP). Finally, we conclude our study with an in vivo analysis of mRNA expression, biodistribution, and tolerability of the TULEP platform in mice. In presenting these data, we hope that our work highlights the utility of TULEPs for tunable and effective mRNA delivery while more broadly highlighting the utility of considering oxygen levels when developing mRNA delivery platforms.
Collapse
Affiliation(s)
- Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
49
|
Banda A, Impomeni O, Singh A, Baloch AR, Hu W, Jaijyan DK. Precision in Action: The Role of Clustered Regularly Interspaced Short Palindromic Repeats/Cas in Gene Therapies. Vaccines (Basel) 2024; 12:636. [PMID: 38932365 PMCID: PMC11209408 DOI: 10.3390/vaccines12060636] [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: 04/14/2024] [Revised: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-associated enzyme-CAS holds great promise for treating many uncured human diseases and illnesses by precisely correcting harmful point mutations and disrupting disease-causing genes. The recent Food and Drug Association (FDA) approval of the first CRISPR-based gene therapy for sickle cell anemia marks the beginning of a new era in gene editing. However, delivering CRISPR specifically into diseased cells in vivo is a significant challenge and an area of intense research. The identification of new CRISPR/Cas variants, particularly ultra-compact CAS systems with robust gene editing activities, paves the way for the low-capacity delivery vectors to be used in gene therapies. CRISPR/Cas technology has evolved beyond editing DNA to cover a wide spectrum of functionalities, including RNA targeting, disease diagnosis, transcriptional/epigenetic regulation, chromatin imaging, high-throughput screening, and new disease modeling. CRISPR/Cas can be used to engineer B-cells to produce potent antibodies for more effective vaccines and enhance CAR T-cells for the more precise and efficient targeting of tumor cells. However, CRISPR/Cas technology has challenges, including off-target effects, toxicity, immune responses, and inadequate tissue-specific delivery. Overcoming these challenges necessitates the development of a more effective and specific CRISPR/Cas delivery system. This entails strategically utilizing specific gRNAs in conjunction with robust CRISPR/Cas variants to mitigate off-target effects. This review seeks to delve into the intricacies of the CRISPR/Cas mechanism, explore progress in gene therapies, evaluate gene delivery systems, highlight limitations, outline necessary precautions, and scrutinize the ethical considerations associated with its application.
Collapse
Affiliation(s)
- Amrutha Banda
- Department of Biology, The College of New Jersey, Ewing Township, NJ 08618, USA
| | - Olivia Impomeni
- Department of Biology, The College of New Jersey, Ewing Township, NJ 08618, USA
| | - Aparana Singh
- Department of Chemistry, National Institute of Technology Agartala, Agartala 799046, India;
| | - Abdul Rasheed Baloch
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Wenhui Hu
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Dabbu Kumar Jaijyan
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23284, USA;
| |
Collapse
|
50
|
Jagannathan JR, Ma Y, Curole BJ, Grayson SM, Fenton OS, Leibfarth FA. Regioselective Palladium-Catalyzed Chain-Growth Allylic Amination Polymerization of Vinyl Aziridines. J Am Chem Soc 2024; 146:15264-15274. [PMID: 38801413 PMCID: PMC11977030 DOI: 10.1021/jacs.4c02599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Organometallic-mediated chain growth polymerization of readily accessible chemical building blocks is responsible for important commercial and technological advances in polymer science, but the incorporation of heteroatoms into the polymer backbone through these mechanisms remains a challenge. Transition metal π-allyl complexes are well-developed organometallic intermediates for carbon-heteroatom bond formation in small-molecule catalysis yet remain underexplored in polymer science. Here, we developed a regioselective palladium-phosphoramidite-catalyzed chain-growth allylic amination polymerization of vinyl aziridines for the synthesis of novel nitrogen-rich polymers via ambiphilic π-allyl complexes. The polymerization accessed a linear microstructure with four carbons between each nitrogen, which is challenging to achieve through other chain-growth polymerization approaches. The highly regioselective allylic amination polymerization demonstrated the characteristics of a controlled polymerization and was able to achieve molar masses exceeding 20 kg mol-1 with low dispersities (D̵ < 1.3). The identification of the polymer structure and well-defined chain ends were supported by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, and chain extension experiments demonstrate opportunities for building more complex materials from this method. A Hammett study was performed to understand the role of the catalyst and monomer structure on regioselectivity, and the data supported a mechanism wherein regioselectivity was primarily controlled by the ligand-metal complex. Postpolymerization desulfonylation provided access to a novel polyamine that demonstrated broad anticancer activity in vitro, which highlights the benefits of unlocking novel polyamine microstructures through regioselective chain-growth allylic amination polymerization.
Collapse
Affiliation(s)
- Jake R. Jagannathan
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Brennan J. Curole
- Department of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Scott M. Grayson
- Department of Chemistry, Tulane University, New Orleans, Louisiana 70118, United States
| | - Owen S. Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Frank A. Leibfarth
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|