1
|
Li J, Sun S, Zhu D, Mei X, Lyu Y, Huang K, Li Y, Liu S, Wang Z, Hu S, Lutz HJ, Popowski KD, Dinh PUC, Butte AJ, Cheng K. Inhalable Stem Cell Exosomes Promote Heart Repair After Myocardial Infarction. Circulation 2024; 150:710-723. [PMID: 39186525 PMCID: PMC11349039 DOI: 10.1161/circulationaha.123.065005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 03/25/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Exosome therapy shows potential for cardiac repair after injury. However, intrinsic challenges such as short half-life and lack of clear targets hinder the clinical feasibility. Here, we report a noninvasive and repeatable method for exosome delivery through inhalation after myocardial infarction (MI), which we called stem cell-derived exosome nebulization therapy (SCENT). METHODS Stem cell-derived exosomes were characterized for size distribution and surface markers. C57BL/6 mice with MI model received exosome inhalation treatment through a nebulizer for 7 consecutive days. Echocardiographies were performed to monitor cardiac function after SCENT, and histological analysis helped with the investigation of myocardial repair. Single-cell RNA sequencing of the whole heart was performed to explore the mechanism of action by SCENT. Last, the feasibility, efficacy, and general safety of SCENT were demonstrated in a swine model of MI, facilitated by 3-dimensional cardiac magnetic resonance imaging. RESULTS Recruitment of exosomes to the ischemic heart after SCENT was detected by ex vivo IVIS imaging and fluorescence microscopy. In a mouse model of MI, SCENT ameliorated cardiac repair by improving left ventricular function, reducing fibrotic tissue, and promoting cardiomyocyte proliferation. Mechanistic studies using single-cell RNA sequencing of mouse heart after SCENT revealed a downregulation of Cd36 in endothelial cells (ECs). In an EC-Cd36fl/- conditional knockout mouse model, the inhibition of CD36, a fatty acid transporter in ECs, led to a compensatory increase in glucose utilization in the heart and higher ATP generation, which enhanced cardiac contractility. In pigs, cardiac magnetic resonance imaging showed an enhanced ejection fraction (Δ=11.66±5.12%) and fractional shortening (Δ=5.72±2.29%) at day 28 after MI by SCENT treatment compared with controls, along with reduced infarct size and thickened ventricular wall. CONCLUSIONS In both rodent and swine models, our data proved the feasibility, efficacy, and general safety of SCENT treatment against acute MI injury, laying the groundwork for clinical investigation. Moreover, the EC-Cd36fl/- mouse model provides the first in vivo evidence showing that conditional EC-CD36 knockout can ameliorate cardiac injury. Our study introduces a noninvasive treatment option for heart disease and identifies new potential therapeutic targets.
Collapse
Affiliation(s)
- Junlang Li
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University (J.L., Y.L., Y.L., Z.W.)
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (J.L., Y.L., K.H., Y.L., Z.W., H.J.L., K.D.P., P.-U.C.D.)
- Xsome Biotech Inc, Raleigh, NC (J.L.)
| | - Shenghuan Sun
- Bakar Computational Health Sciences Institute, University of California, San Francisco (S.S., A.J.B.)
| | - Dashuai Zhu
- Department of Biomedical Engineering (D.Z., S.L., S.H., K.C.), Columbia University, New York, NY
| | - Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA (X.M.)
| | - Yongbo Lyu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University (J.L., Y.L., Y.L., Z.W.)
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (J.L., Y.L., K.H., Y.L., Z.W., H.J.L., K.D.P., P.-U.C.D.)
| | - Ke Huang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (J.L., Y.L., K.H., Y.L., Z.W., H.J.L., K.D.P., P.-U.C.D.)
| | - Yuan Li
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University (J.L., Y.L., Y.L., Z.W.)
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (J.L., Y.L., K.H., Y.L., Z.W., H.J.L., K.D.P., P.-U.C.D.)
| | - Shuo Liu
- Department of Biomedical Engineering (D.Z., S.L., S.H., K.C.), Columbia University, New York, NY
| | - Zhenzhen Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University (J.L., Y.L., Y.L., Z.W.)
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (J.L., Y.L., K.H., Y.L., Z.W., H.J.L., K.D.P., P.-U.C.D.)
| | - Shiqi Hu
- Department of Biomedical Engineering (D.Z., S.L., S.H., K.C.), Columbia University, New York, NY
| | - Halle J Lutz
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (J.L., Y.L., K.H., Y.L., Z.W., H.J.L., K.D.P., P.-U.C.D.)
| | - Kristen D Popowski
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (J.L., Y.L., K.H., Y.L., Z.W., H.J.L., K.D.P., P.-U.C.D.)
| | - Phuong-Uyen C Dinh
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh (J.L., Y.L., K.H., Y.L., Z.W., H.J.L., K.D.P., P.-U.C.D.)
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California, San Francisco (S.S., A.J.B.)
| | - Ke Cheng
- Department of Biomedical Engineering (D.Z., S.L., S.H., K.C.), Columbia University, New York, NY
- Herbert Irving Comprehensive Cancer Center (K.C.), Columbia University, New York, NY
| |
Collapse
|
2
|
Seidl LL, Moog R, Graeser KA. Antisense oligonucleotides and their technical suitability to nebulization. Int J Pharm 2024; 661:124390. [PMID: 38936443 DOI: 10.1016/j.ijpharm.2024.124390] [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: 01/17/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
In vivo studies investigating the inhalative efficacy of biotherapeutics, such as nucleic acids, usually do not perform an aerosolization step, rather the solution is directly administered into the lungs e.g. intratracheally. In addition, there is currently very little information on the behavior of nucleic acid solutions when subjected to the physical stress of the nebulization process. In this study, the aim was to assess the technical suitability of Locked Nucleic Acids (LNAs), as a model antisense oligonucleotide, towards nebulization using two commercially available nebulizers. A jet nebulizer (Pari LC Plus) and a vibrating mesh nebulizer (Aerogen Solo) were employed and solutions of five different LNAs investigated in terms of their physical and chemical stability to nebulization and the quality of the generated aerosols. The aerosol properties of the Aerogen Solo were mainly influenced by the viscosity of the solutions with the output rate and the droplet size decreasing with increasing viscosity. The Pari LC Plus was less susceptible to viscosity and overall the droplet size was smaller. The LNAs tolerated both nebulization processes and the integrity of the molecules was shown. Chemical stability of the molecules from the Aerogen Solo was confirmed, whereas aerosol generation with the Pari LC Plus jet nebulizer led to a slight increase of phosphodiester groups in a fully phosphorothiolated backbone of the LNAs. Overall, it could be shown that nebulization of different LNAs is possible and inhalation can therefore be considered a potential route of administration.
Collapse
Affiliation(s)
- Leonardo L Seidl
- Roche Pharma Research and Early Development, Therapeutic Modalities, pCMC, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland; Technical University of Munich, TUM School of Natural Sciences, Boltzmannstr. 10, 85748 Garching, Germany
| | - Regina Moog
- Roche Pharma Research and Early Development, Therapeutic Modalities, pCMC, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Kirsten A Graeser
- Roche Pharma Research and Early Development, Therapeutic Modalities, pCMC, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland.
| |
Collapse
|
3
|
Huang Y, Zhang J, Wang X, Jing H, Li H. Aerosol Inhalation of Gene Delivery Therapy for Pulmonary Diseases. Biomolecules 2024; 14:904. [PMID: 39199292 PMCID: PMC11352762 DOI: 10.3390/biom14080904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
Gene delivery therapy has emerged as a popular approach for the treatment of various diseases. However, it still poses the challenges of accumulation in target sites and reducing off-target effects. Aerosol gene delivery for the treatment of pulmonary diseases has the advantages of high lung accumulation, specific targeting and fewer systemic side effects. However, the key challenge is selecting the appropriate formulation for aerosol gene delivery that can overcome physiological barriers. There are numerous existing gene carriers under study, including viral vectors and non-viral vectors. With the development of biomaterials, more biocompatible substances have applied gene delivery via inhalation. Furthermore, many types of genes can be delivered through aerosol inhalation, such as DNA, mRNA, siRNA and CRISPR/Cas9. Aerosol delivery of different types of genes has proven to be efficient in the treatment of many diseases such as SARS-CoV-2, cystic fibrosis and lung cancer. In this paper, we provide a comprehensive review of the ongoing research on aerosol gene delivery therapy, including the basic respiratory system, different types of gene carriers, different types of carried genes and clinical applications.
Collapse
Affiliation(s)
| | | | | | - Hui Jing
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.H.); (J.Z.); (X.W.)
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.H.); (J.Z.); (X.W.)
| |
Collapse
|
4
|
Li J, Luo L, He J, Yu J, Li X, Shen X, Zhang J, Li S, Karp JM, Kuai R. A Virus-Inspired Inhalable Liponanogel Induces Potent Antitumor Immunity and Regression in Metastatic Lung Tumors. Cancer Res 2024; 84:2352-2363. [PMID: 38718316 PMCID: PMC11247319 DOI: 10.1158/0008-5472.can-23-3414] [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: 10/31/2023] [Revised: 02/27/2024] [Accepted: 05/02/2024] [Indexed: 07/16/2024]
Abstract
Pulmonary delivery of immunostimulatory agents such as poly(I:C) to activate double-stranded RNA sensors MDA5 and RIG-I within lung-resident antigen-presenting cells is a potential strategy to enhance antitumor immunity by promoting type I interferon secretion. Nevertheless, following pulmonary delivery, poly(I:C) suffers from rapid degradation and poor endosomal escape, thus limiting its potency. Inspired by the structure of a virus that utilizes internal viral proteins to tune the loading and cytosolic delivery of viral nucleic acids, we developed a liponanogel (LNG)-based platform to overcome the delivery challenges of poly(I:C). The LNG comprised an anionic polymer hyaluronic acid-based nanogel core coated by a lipid shell, which served as a protective layer to stabilize the nanogel core in the lungs. The nanogel core was protonated within acidic endosomes to enhance the endosomal membrane permeability and cytosolic delivery of poly(I:C). After pulmonary delivery, LNG-poly(I:C) induced 13.7-fold more IFNβ than poly(I:C) alone and two-fold more than poly(I:C) loaded in the state-of-art lipid nanoparticles [LNP-poly(I:C)]. Additionally, LNG-poly(I:C) induced more potent CD8+ T-cell immunity and stronger therapeutic effects than LNP-poly(I:C). The combination of LNG-poly(I:C) and PD-L1 targeting led to regression of established lung metastases. Due to the ease of manufacturing and the high biocompatibility of LNG, pulmonary delivery of LNG may be broadly applicable to the treatment of different lung tumors and may spur the development of innovative strategies for cancer immunotherapy. Significance: Pulmonary delivery of poly(I:C) with a virus-inspired inhalable liponanogel strongly activates cytosolic MDA5 and RIG-I and stimulates antitumor immunity, representing a promising strategy for safe and effective treatment of metastatic lung tumors.
Collapse
Affiliation(s)
- Junyao Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Lanqing Luo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Jinchao Yu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Xueying Shen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Junxia Zhang
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China.
| | - Sai Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China.
| | - Jeffrey M. Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts.
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, Massachusetts.
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts.
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| |
Collapse
|
5
|
Woodward IR, Fromen CA. Recent Developments in Aerosol Pulmonary Drug Delivery: New Technologies, New Cargos, and New Targets. Annu Rev Biomed Eng 2024; 26:307-330. [PMID: 38424089 PMCID: PMC11222059 DOI: 10.1146/annurev-bioeng-110122-010848] [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] [Indexed: 03/02/2024]
Abstract
There is nothing like a global pandemic to motivate the need for improved respiratory treatments and mucosal vaccines. Stimulated by the COVID-19 pandemic, pulmonary aerosol drug delivery has seen a flourish of activity, building on the prior decades of innovation in particle engineering, inhaler device technologies, and clinical understanding. As such, the field has expanded into new directions and is working toward the efficient delivery of increasingly complex cargos to address a wider range of respiratory diseases. This review seeks to highlight recent innovations in approaches to personalize inhalation drug delivery, deliver complex cargos, and diversify the targets treated and prevented through pulmonary drug delivery. We aim to inform readers of the emerging efforts within the field and predict where future breakthroughs are expected to impact the treatment of respiratory diseases.
Collapse
Affiliation(s)
- Ian R Woodward
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA;
| | - Catherine A Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA;
| |
Collapse
|
6
|
Wang Q, Bu C, Dai Q, Chen J, Zhang R, Zheng X, Ren H, Xin X, Li X. Recent Progress in Nucleic Acid Pulmonary Delivery toward Overcoming Physiological Barriers and Improving Transfection Efficiency. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309748. [PMID: 38460157 PMCID: PMC11095210 DOI: 10.1002/advs.202309748] [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: 12/12/2023] [Revised: 02/04/2024] [Indexed: 03/11/2024]
Abstract
Pulmonary delivery of therapeutic agents has been considered the desirable administration route for local lung disease treatment. As the latest generation of therapeutic agents, nucleic acid has been gradually developed as gene therapy for local diseases such as asthma, chronic obstructive pulmonary diseases, and lung fibrosis. The features of nucleic acid, specific physiological structure, and pathophysiological barriers of the respiratory tract have strongly affected the delivery efficiency and pulmonary bioavailability of nucleic acid, directly related to the treatment outcomes. The development of pharmaceutics and material science provides the potential for highly effective pulmonary medicine delivery. In this review, the key factors and barriers are first introduced that affect the pulmonary delivery and bioavailability of nucleic acids. The advanced inhaled materials for nucleic acid delivery are further summarized. The recent progress of platform designs for improving the pulmonary delivery efficiency of nucleic acids and their therapeutic outcomes have been systematically analyzed, with the application and the perspectives of advanced vectors for pulmonary gene delivery.
Collapse
Affiliation(s)
- Qiyue Wang
- School of Pharmaceutical ScienceNanjing Tech UniversityNanjing211816China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparation and ExcipientsNanjing210009China
| | - Chaozhi Bu
- Wuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxi214002China
| | - Qihao Dai
- School of Pharmaceutical ScienceNanjing Tech UniversityNanjing211816China
| | - Jinhua Chen
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparation and ExcipientsNanjing210009China
- Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Ruitao Zhang
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparation and ExcipientsNanjing210009China
- Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Xiaomin Zheng
- Wuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxi214002China
| | - Hao Ren
- School of Pharmaceutical ScienceNanjing Tech UniversityNanjing211816China
| | - Xiaofei Xin
- Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of PharmaceuticsChina Pharmaceutical UniversityNanjing210009China
| | - Xueming Li
- School of Pharmaceutical ScienceNanjing Tech UniversityNanjing211816China
| |
Collapse
|
7
|
Teng M, Xia ZJ, Lo N, Daud K, He HH. Assembling the RNA therapeutics toolbox. MEDICAL REVIEW (2021) 2024; 4:110-128. [PMID: 38680684 PMCID: PMC11046573 DOI: 10.1515/mr-2023-0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/29/2024] [Indexed: 05/01/2024]
Abstract
From the approval of COVID-19 mRNA vaccines to the 2023 Nobel Prize awarded for nucleoside base modifications, RNA therapeutics have entered the spotlight and are transforming drug development. While the term "RNA therapeutics" has been used in various contexts, this review focuses on treatments that utilize RNA as a component or target RNA for therapeutic effects. We summarize the latest advances in RNA-targeting tools and RNA-based technologies, including but not limited to mRNA, antisense oligos, siRNAs, small molecules and RNA editors. We focus on the mechanisms of current FDA-approved therapeutics but also provide a discussion on the upcoming workforces. The clinical utility of RNA-based therapeutics is enabled not only by the advances in RNA technologies but in conjunction with the significant improvements in chemical modifications and delivery platforms, which are also briefly discussed in the review. We summarize the latest RNA therapeutics based on their mechanisms and therapeutic effects, which include expressing proteins for vaccination and protein replacement therapies, degrading deleterious RNA, modulating transcription and translation efficiency, targeting noncoding RNAs, binding and modulating protein activity and editing RNA sequences and modifications. This review emphasizes the concept of an RNA therapeutic toolbox, pinpointing the readers to all the tools available for their desired research and clinical goals. As the field advances, the catalog of RNA therapeutic tools continues to grow, further allowing researchers to combine appropriate RNA technologies with suitable chemical modifications and delivery platforms to develop therapeutics tailored to their specific clinical challenges.
Collapse
Affiliation(s)
- Mona Teng
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ziting Judy Xia
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Nicholas Lo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Kashif Daud
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Housheng Hansen He
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
8
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
9
|
Xiao H, Amarsaikhan O, Zhao Y, Yu X, Hu X, Han S, Chaolumen, Baigude H. Astrocyte-targeted siRNA delivery by adenosine-functionalized LNP in mouse TBI model. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102065. [PMID: 38028196 PMCID: PMC10661454 DOI: 10.1016/j.omtn.2023.102065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023]
Abstract
Traumatic brain injury (TBI) induces pro-inflammatory polarization of astrocytes and causes secondary disruption of the blood-brain barrier (BBB) and brain damage. Herein, we report a successful astrocyte-targeted delivery of small interfering RNA (siRNA) by ligand functionalized lipid nanoparticles (LNPs) formulated from adenosine-conjugated lipids and a novel ionizable lipid (denoted by Ad4 LNPs). Systemic administration of Ad4 LNPs carrying siRNA against TLR4 to the mice TBI model resulted in the specific internalization of the LNPs by astrocytes in the vicinity of damaged brain tissue. A substantial knockdown of TLR4 at both mRNA and protein levels in the brain was observed, which led to a significant decrease of key pro-inflammatory cytokines and an increase of key anti-inflammatory cytokines in serum. Dye leakage measurement suggested that the Ad4-LNP-mediated knockdown of TLR4 attenuated the TBI-induced BBB disruption. Together, our data suggest that Ad4 LNP is a promising vehicle for astrocyte-specific delivery of nucleic acid therapeutics.
Collapse
Affiliation(s)
- Hai Xiao
- Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Odmaa Amarsaikhan
- Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Yunwang Zhao
- Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Xiang Yu
- Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Xin Hu
- Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Shuqin Han
- Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Chaolumen
- Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| | - Huricha Baigude
- Inner Mongolia Key Laboratory of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P.R. China
| |
Collapse
|
10
|
Kevadiya BD, Islam F, Deol P, Zaman LA, Mosselhy DA, Ashaduzzaman M, Bajwa N, Routhu NK, Singh PA, Dawre S, Vora LK, Nahid S, Mathur D, Nayan MU, Baldi A, Kothari R, Patel TA, Madan J, Gounani Z, Bariwal J, Hettie KS, Gendelman HE. Delivery of gene editing therapeutics. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 54:102711. [PMID: 37813236 PMCID: PMC10843524 DOI: 10.1016/j.nano.2023.102711] [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: 04/24/2023] [Revised: 08/31/2023] [Accepted: 09/15/2023] [Indexed: 10/11/2023]
Abstract
For the past decades, gene editing demonstrated the potential to attenuate each of the root causes of genetic, infectious, immune, cancerous, and degenerative disorders. More recently, Clustered Regularly Interspaced Short Palindromic Repeats-CRISPR-associated protein 9 (CRISPR-Cas9) editing proved effective for editing genomic, cancerous, or microbial DNA to limit disease onset or spread. However, the strategies to deliver CRISPR-Cas9 cargos and elicit protective immune responses requires safe delivery to disease targeted cells and tissues. While viral vector-based systems and viral particles demonstrate high efficiency and stable transgene expression, each are limited in their packaging capacities and secondary untoward immune responses. In contrast, the nonviral vector lipid nanoparticles were successfully used for as vaccine and therapeutic deliverables. Herein, we highlight each available gene delivery systems for treating and preventing a broad range of infectious, inflammatory, genetic, and degenerative diseases. STATEMENT OF SIGNIFICANCE: CRISPR-Cas9 gene editing for disease treatment and prevention is an emerging field that can change the outcome of many chronic debilitating disorders.
Collapse
Affiliation(s)
- Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| | - Farhana Islam
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| | - Pallavi Deol
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Institute of Modeling Collaboration and Innovation and Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA.
| | - Lubaba A Zaman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| | - Dina A Mosselhy
- Department of Virology, Faculty of Medicine, University of Helsinki, P.O. Box 21, 00014 Helsinki, Finland; Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, 00014 Helsinki, Finland; Microbiological Unit, Fish Diseases Department, Animal Health Research Institute, ARC, Dokki, Giza 12618, Egypt.
| | - Md Ashaduzzaman
- Department of Computer Science, University of Nebraska Omaha, Omaha, NE 68182, USA.
| | - Neha Bajwa
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Nanda Kishore Routhu
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| | - Preet Amol Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India; Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab.
| | - Shilpa Dawre
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKMs, NMIMS, Babulde Banks of Tapi River, MPTP Park, Mumbai-Agra Road, Shirpur, Maharashtra, 425405, India.
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom.
| | - Sumaiya Nahid
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| | | | - Mohammad Ullah Nayan
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| | - Ashish Baldi
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India; Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab.
| | - Ramesh Kothari
- Department of Biosciences, Saurashtra University, Rajkot 360005, Gujarat, India.
| | - Tapan A Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-NIPER, Hyderabad 500037, Telangana, India.
| | - Zahra Gounani
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, 00790 Helsinki, Finland.
| | - Jitender Bariwal
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, School of Medicine, 3601 4th Street, Lubbock, TX 79430-6551, USA.
| | - Kenneth S Hettie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Department of Otolaryngology - Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
11
|
Li J, Wang H. Selective organ targeting nanoparticles: from design to clinical translation. NANOSCALE HORIZONS 2023; 8:1155-1173. [PMID: 37427677 DOI: 10.1039/d3nh00145h] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Targeting nanoparticle is a very promising therapeutic approach that can precisely target specific sites to treat diseases. Research on nanoscale drug delivery systems has made great progress in the past few years, making targeting nanoparticles a promising prospect. However, selective targeting nanoparticles designed for specific organs still face several challenges, one of which is the unknown fate of nanoparticles in vivo. This review starts with the in vivo journey of nanoparticles and describes the biological barriers and some targeting strategies for nanoparticles to target specific organs. Then, through the collection of literature in recent years, the design of selective targeting nanoparticles for various organs is illustrated, which provides a reference strategy for people to study the design of selective organ targeting nanoparticles. Ultimately, the prospect and challenge of selective organ targeting nanoparticles are discussed by collecting the data of clinical trials and marketed drugs.
Collapse
Affiliation(s)
- Jian Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
12
|
van Rijn CJM, Vlaming KE, Bem RA, Dekker RJ, Poortinga A, Breit T, van Leeuwen S, Ensink WA, van Wijnbergen K, van Hamme JL, Bonn D, Geijtenbeek TBH. Low energy nebulization preserves integrity of SARS-CoV-2 mRNA vaccines for respiratory delivery. Sci Rep 2023; 13:8851. [PMID: 37258559 DOI: 10.1038/s41598-023-35872-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023] Open
Abstract
Nebulization of mRNA therapeutics can be used to directly target the respiratory tract. A promising prospect is that mucosal administration of lipid nanoparticle (LNP)-based mRNA vaccines may lead to a more efficient protection against respiratory viruses. However, the nebulization process can rupture the LNP vehicles and degrade the mRNA molecules inside. Here we present a novel nebulization method able to preserve substantially the integrity of vaccines, as tested with two SARS-CoV-2 mRNA vaccines. We compare the new method with well-known nebulization methods used for medical respiratory applications. We find that a lower energy level in generating LNP droplets using the new nebulization method helps safeguard the integrity of the LNP and vaccine. By comparing nebulization techniques with different energy dissipation levels we find that LNPs and mRNAs can be kept largely intact if the energy dissipation remains below a threshold value, for LNP integrity 5-10 J/g and for mRNA integrity 10-20 J/g for both vaccines.
Collapse
Affiliation(s)
- Cees J M van Rijn
- van der Waals-Zeeman Institute, Institute of Physics, University of Amsterdam, Amsterdam, The Netherlands
| | - Killian E Vlaming
- Department of Experimental Immunology, Amsterdam University Medical Centers, Meibergdreef 9, Amsterdam, The Netherlands
- Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Reinout A Bem
- Pediatric Intensive Care Unit, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Rob J Dekker
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Albert Poortinga
- van der Waals-Zeeman Institute, Institute of Physics, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo Breit
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Selina van Leeuwen
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Wim A Ensink
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Kelly van Wijnbergen
- Department of Experimental Immunology, Amsterdam University Medical Centers, Meibergdreef 9, Amsterdam, The Netherlands
- Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - John L van Hamme
- Department of Experimental Immunology, Amsterdam University Medical Centers, Meibergdreef 9, Amsterdam, The Netherlands
- Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Daniel Bonn
- van der Waals-Zeeman Institute, Institute of Physics, University of Amsterdam, Amsterdam, The Netherlands.
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Centers, Meibergdreef 9, Amsterdam, The Netherlands
- Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Ferguson LT, Ma X, Myerson JW, Wu J, Glassman PM, Zamora ME, Hood ED, Zaleski M, Shen M, Essien EO, Shuvaev VV, Brenner JS. Mechanisms by Which Liposomes Improve Inhaled Drug Delivery for Alveolar Diseases. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200106. [PMID: 37266328 PMCID: PMC10231510 DOI: 10.1002/anbr.202200106] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/23/2022] [Indexed: 01/29/2023] Open
Abstract
Diseases of the pulmonary alveolus, such as pulmonary fibrosis, are leading causes of morbidity and mortality, but exceedingly few drugs are developed for them. A major reason for this gap is that after inhalation, drugs are quickly whisked away from alveoli due to their high perfusion. To solve this problem, the mechanisms by which nano-scale drug carriers dramatically improve lung pharmacokinetics using an inhalable liposome formulation containing nintedanib, an antifibrotic for pulmonary fibrosis, are studied. Direct instillation of liposomes in murine lung increases nintedanib's total lung delivery over time by 8000-fold and lung half life by tenfold, compared to oral nintedanib. Counterintuitively, it is shown that pulmonary surfactant neither lyses nor aggregates the liposomes. Instead, each lung compartment (alveolar fluid, alveolar leukocytes, and parenchyma) elutes liposomes over 24 h, likely serving as "drug depots." After deposition in the surfactant layer, liposomes are transferred over 3-6 h to alveolar leukocytes (which take up a surprisingly minor 1-5% of total lung dose instilled) in a nonsaturable fashion. Further, all cell layers of the lung parenchyma take up liposomes. These and other mechanisms elucidated here should guide engineering of future inhaled nanomedicine for alveolar diseases.
Collapse
Affiliation(s)
- Laura T. Ferguson
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Xiaonan Ma
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jacob W. Myerson
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jichuan Wu
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Patrick M. Glassman
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Marco E. Zamora
- School of Biomedical Engineering, Science, and Health SystemsDrexel UniversityPhiladelphiaPA19104USA
| | - Elizabeth D. Hood
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Michael Zaleski
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Mengwen Shen
- Emergency Medicine DepartmentYueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese Medicine200437ShanghaiChina
- Department of MicrobiologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Eno-Obong Essien
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Vladimir V. Shuvaev
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Jacob S. Brenner
- Department of MedicinePulmonary, Allergy, and Critical Care DivisionPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Systems Pharmacology and Translational TherapeuticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Penn-CHOP Lung Biology InstitutePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| |
Collapse
|
14
|
Popowski KD, López de Juan Abad B, George A, Silkstone D, Belcher E, Chung J, Ghodsi A, Lutz H, Davenport J, Flanagan M, Piedrahita J, Dinh PUC, Cheng K. Inhalable exosomes outperform liposomes as mRNA and protein drug carriers to the lung. EXTRACELLULAR VESICLE 2022; 1:100002. [PMID: 36523538 PMCID: PMC9213043 DOI: 10.1016/j.vesic.2022.100002] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/09/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023]
Abstract
Respiratory diseases are among the leading causes of morbidity and mortality worldwide, coupled with the ongoing coronavirus disease 2019 (COVID-19) pandemic. mRNA lipid nanoparticle (LNP) vaccines have been developed, but their intramuscular delivery limits pulmonary bioavailability. Inhalation of nanoparticle therapeutics offers localized drug delivery that minimizes off targeted adverse effects and has greater patient compliance. However, LNP platforms require extensive reformulation for inhaled delivery. Lung-derived extracellular vesicles (Lung-Exo) offer a biological nanoparticle alternative that is naturally optimized for mRNA translation and delivery to pulmonary cells. We compared the biodistribution of Lung-Exo against commercially standard biological extracellular vesicles (HEK-Exo) and LNPs (Lipo), where Lung-Exo exhibited superior mRNA and protein cargo distribution to and retention in the bronchioles and parenchyma following nebulization administration. This suggests that inhaled Lung-Exo can deliver mRNA and protein drugs with enhanced pulmonary bioavailability and therapeutic efficacy.
Collapse
Affiliation(s)
- Kristen D Popowski
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Arianna George
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA
| | - Dylan Silkstone
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, 27607/27599, USA
| | - Elizabeth Belcher
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, 27607/27599, USA
| | - Jaewook Chung
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
| | - Asma Ghodsi
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Halle Lutz
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
| | - Jada Davenport
- Department of Animal Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Mallory Flanagan
- Department of Animal Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Jorge Piedrahita
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
| | - Phuong-Uyen C Dinh
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, 27607/27599, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
15
|
Zadory M, Lopez E, Babity S, Gravel SP, Brambilla D. Current knowledge on the tissue distribution of mRNA nanocarriers for therapeutic protein expression. Biomater Sci 2022; 10:6077-6115. [PMID: 36097955 DOI: 10.1039/d2bm00859a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Exogenously delivered mRNA-based drugs are emerging as a new class of therapeutics with the potential to treat several diseases. Over the last decade, advancements in the design of non-viral delivery tools have enabled mRNA to be evaluated for several therapeutic purposes including protein replacement therapies, gene editing, and vaccines. However, in vivo delivery of mRNA to targeted organs and cells remains a critical challenge. Evaluation of the biodistribution of mRNA vehicles is of utmost importance for the development of effective pharmaceutical candidates. In this review, we discuss the recent advances in the design of nanoparticles loaded with mRNA and extrapolate the key factors influencing their biodistribution following administration. Finally, we highlight the latest developments in the preclinical and clinical translation of mRNA therapeutics for protein supplementation therapy.
Collapse
Affiliation(s)
- Matthias Zadory
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Elliot Lopez
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Samuel Babity
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Simon-Pierre Gravel
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| | - Davide Brambilla
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, Québec, Canada, H3T 1J4.
| |
Collapse
|
16
|
Leong EWX, Ge R. Lipid Nanoparticles as Delivery Vehicles for Inhaled Therapeutics. Biomedicines 2022; 10:2179. [PMID: 36140280 PMCID: PMC9496059 DOI: 10.3390/biomedicines10092179] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Lipid nanoparticles (LNPs) have emerged as a powerful non-viral carrier for drug delivery. With the prevalence of respiratory diseases, particularly highlighted by the current COVID-19 pandemic, investigations into applying LNPs to deliver inhaled therapeutics directly to the lungs are underway. The progress in LNP development as well as the recent pre-clinical studies in three main classes of inhaled encapsulated drugs: small molecules, nucleic acids and proteins/peptides will be discussed. The advantages of the pulmonary drug delivery system such as reducing systemic toxicity and enabling higher local drug concentration in the lungs are evaluated together with the challenges and design considerations for improved formulations. This review provides a perspective on the future prospects of LNP-mediated delivery of inhaled therapeutics for respiratory diseases.
Collapse
Affiliation(s)
| | - Ruowen Ge
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117558, Singapore
| |
Collapse
|
17
|
Advancements in Particle Engineering for Inhalation Delivery of Small Molecules and Biotherapeutics. Pharm Res 2022; 39:3047-3061. [PMID: 36071354 PMCID: PMC9451127 DOI: 10.1007/s11095-022-03363-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/06/2022] [Indexed: 12/27/2022]
Abstract
Dry powder inhalation formulations have become increasingly popular for local and systemic delivery of small molecules and biotherapeutics. Powder formulations provide distinct advantages over liquid formulations such as elimination of cold chain due to room temperature stability, improved portability, and the potential for increasing patient adherence. To become a viable product, it is essential to develop formulations that are stable (physically, chemically and/or biologically) and inhalable over the shelf-life. Physical particulate properties such as particle size, morphology and density, as well as chemical properties can significantly impact aerosol performance of the powder. This review will cover these critical attributes that can be engineered to enhance the dispersibility of inhalation powder formulations. Challenges in particle engineering for biotherapeutics will be assessed, followed by formulation strategies for overcoming the hurdles. Finally, the review will discuss recent examples of successful dry powder biotherapeutic formulations for inhalation delivery that have been clinically assessed.
Collapse
|