201
|
Weng Y, Xiao H, Zhang J, Liang XJ, Huang Y. RNAi therapeutic and its innovative biotechnological evolution. Biotechnol Adv 2019; 37:801-825. [PMID: 31034960 DOI: 10.1016/j.biotechadv.2019.04.012] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 04/09/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023]
Abstract
Recently, United States Food and Drug Administration (FDA) and European Commission (EC) approved Alnylam Pharmaceuticals' RNA interference (RNAi) therapeutic, ONPATTRO™ (Patisiran), for the treatment of the polyneuropathy of hereditary transthyretin-mediated (hATTR) amyloidosis in adults. This is the first RNAi therapeutic all over the world, as well as the first FDA-approved treatment for this indication. As a milestone event in RNAi pharmaceutical industry, it means, for the first time, people have broken through all development processes for RNAi drugs from research to clinic. With this achievement, RNAi approval may soar in the coming years. In this paper, we introduce the basic information of ONPATTRO and the properties of RNAi and nucleic acid therapeutics, update the clinical and preclinical development activities, review its complicated development history, summarize the key technologies of RNAi at early stage, and discuss the latest advances in delivery and modification technologies. It provides a comprehensive view and biotechnological insights of RNAi therapy for the broader audiences.
Collapse
Affiliation(s)
- Yuhua Weng
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, PR China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jinchao Zhang
- College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, PR China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, PR China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, PR China.
| |
Collapse
|
202
|
Li Y, Chakraborty A, Chen J, Xu Q. Combinatorial Library of Light-Cleavable Lipidoid Nanoparticles for Intracellular Drug Delivery. ACS Biomater Sci Eng 2019; 5:2391-2398. [DOI: 10.1021/acsbiomaterials.9b00445] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Anirban Chakraborty
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Jinjin Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
203
|
Kowalski PS, Rudra A, Miao L, Anderson DG. Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery. Mol Ther 2019; 27:710-728. [PMID: 30846391 PMCID: PMC6453548 DOI: 10.1016/j.ymthe.2019.02.012] [Citation(s) in RCA: 621] [Impact Index Per Article: 124.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 12/18/2022] Open
Abstract
mRNA has broad potential as a therapeutic. Current clinical efforts are focused on vaccination, protein replacement therapies, and treatment of genetic diseases. The clinical translation of mRNA therapeutics has been made possible through advances in the design of mRNA manufacturing and intracellular delivery methods. However, broad application of mRNA is still limited by the need for improved delivery systems. In this review, we discuss the challenges for clinical translation of mRNA-based therapeutics, with an emphasis on recent advances in biomaterials and delivery strategies, and we present an overview of the applications of mRNA-based delivery for protein therapy, gene editing, and vaccination.
Collapse
Affiliation(s)
- Piotr S Kowalski
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Arnab Rudra
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Lei Miao
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Anesthesiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
204
|
Paunovska K, Da Silva Sanchez AJ, Sago CD, Gan Z, Lokugamage MP, Islam FZ, Kalathoor S, Krupczak BR, Dahlman JE. Nanoparticles Containing Oxidized Cholesterol Deliver mRNA to the Liver Microenvironment at Clinically Relevant Doses. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807748. [PMID: 30748040 PMCID: PMC6445717 DOI: 10.1002/adma.201807748] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/02/2019] [Indexed: 05/14/2023]
Abstract
Using mRNA to produce therapeutic proteins is a promising approach to treat genetic diseases. However, systemically delivering mRNA to cell types besides hepatocytes remains challenging. Fast identification of nanoparticle delivery (FIND) is a DNA barcode-based system designed to measure how over 100 lipid nanoparticles (LNPs) deliver mRNA that functions in the cytoplasm of target cells in a single mouse. By using FIND to quantify how 75 chemically distinct LNPs delivered mRNA to 28 cell types in vivo, it is found that an LNP formulated with oxidized cholesterol and no targeting ligand delivers Cre mRNA, which edits DNA in hepatic endothelial cells and Kupffer cells at 0.05 mg kg-1 . Notably, the LNP targets liver microenvironmental cells fivefold more potently than hepatocytes. The structure of the oxidized cholesterols added to the LNP is systematically varied to show that the position of the oxidative modification may be important; cholesterols modified on the hydrocarbon tail associated with sterol ring D tend to outperform cholesterols modified on sterol ring B. These data suggest that LNPs formulated with modified cholesterols can deliver gene-editing mRNA to the liver microenvironment at clinically relevant doses.
Collapse
Affiliation(s)
- Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Alejandro J Da Silva Sanchez
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Zubao Gan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Fatima Z Islam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Sujay Kalathoor
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - Brandon R Krupczak
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
| |
Collapse
|
205
|
Dong Y, Siegwart DJ, Anderson DG. Strategies, design, and chemistry in siRNA delivery systems. Adv Drug Deliv Rev 2019; 144:133-147. [PMID: 31102606 DOI: 10.1016/j.addr.2019.05.004] [Citation(s) in RCA: 331] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/03/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022]
Abstract
Emerging therapeutics that utilize RNA interference (RNAi) have the potential to treat broad classes of diseases due to their ability to reversibly silence target genes. In August 2018, the FDA approved the first siRNA therapeutic, called ONPATTRO™ (Patisiran), for the treatment of transthyretin-mediated amyloidosis. This was an important milestone for the field of siRNA delivery that opens the door for additional siRNA drugs. Currently, >20 small interfering RNA (siRNA)-based therapies are in clinical trials for a wide variety of diseases including cancers, genetic disorders, and viral infections. To maximize therapeutic benefits of siRNA-based drugs, a number of chemical strategies have been applied to address issues associated with efficacy, specificity, and safety. This review focuses on the chemical perspectives behind non-viral siRNA delivery systems, including siRNA synthesis, siRNA conjugates, and nanoparticle delivery using nucleotides, lipids, and polymers. Tracing and understanding the chemical development of strategies to make siRNAs into drugs is important to guide development of additional clinical candidates and enable prolonged success of siRNA therapeutics.
Collapse
Affiliation(s)
- Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| | - Daniel J Siegwart
- Simmons Comprehensive Cancer Center, Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| | - Daniel G Anderson
- Deparment of Chemical Engineering, David H. Koch Institute for Integrative Cancer Research, Department of Chemistry, Institute for Medical Engineering and Science, and Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, United States.
| |
Collapse
|
206
|
RNA Circularization Diminishes Immunogenicity and Can Extend Translation Duration In Vivo. Mol Cell 2019; 74:508-520.e4. [PMID: 30902547 DOI: 10.1016/j.molcel.2019.02.015] [Citation(s) in RCA: 242] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/13/2018] [Accepted: 02/11/2019] [Indexed: 12/13/2022]
Abstract
Circular RNAs (circRNAs) are a class of single-stranded RNAs with a contiguous structure that have enhanced stability and a lack of end motifs necessary for interaction with various cellular proteins. Here, we show that unmodified exogenous circRNA is able to bypass cellular RNA sensors and thereby avoid provoking an immune response in RIG-I and Toll-like receptor (TLR) competent cells and in mice. The immunogenicity and protein expression stability of circRNA preparations are found to be dependent on purity, with small amounts of contaminating linear RNA leading to robust cellular immune responses. Unmodified circRNA is less immunogenic than unmodified linear mRNA in vitro, in part due to the evasion of TLR sensing. Finally, we provide the first demonstration to our knowledge of exogenous circRNA delivery and translation in vivo, and we show that circRNA translation is extended in adipose tissue in comparison to unmodified and uridine-modified linear mRNAs.
Collapse
|
207
|
Li B, Zhang X, Dong Y. Nanoscale platforms for messenger RNA delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1530. [PMID: 29726120 PMCID: PMC6443240 DOI: 10.1002/wnan.1530] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/27/2018] [Accepted: 04/01/2018] [Indexed: 12/27/2022]
Abstract
Messenger RNA (mRNA) has become a promising class of drugs for diverse therapeutic applications in the past few years. A series of clinical trials are ongoing or will be initiated in the near future for the treatment of a variety of diseases. Currently, mRNA-based therapeutics mainly focuses on ex vivo transfection and local administration in clinical studies. Efficient and safe delivery of therapeutically relevant mRNAs remains one of the major challenges for their broad applications in humans. Thus, effective delivery systems are urgently needed to overcome this limitation. In recent years, numerous nanoscale biomaterials have been constructed for mRNA delivery in order to protect mRNA from extracellular degradation and facilitate endosomal escape after cellular uptake. Nanoscale platforms have expanded the feasibility of mRNA-based therapeutics, and enabled its potential applications to protein replacement therapy, cancer immunotherapy, therapeutic vaccines, regenerative medicine, and genome editing. This review focuses on recent advances, challenges, and future directions in nanoscale platforms designed for mRNA delivery, including lipid and lipid-derived nanoparticles, polymer-based nanoparticles, protein derivatives mRNA complexes, and other types of nanomaterials. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Biology-Inspired Nanomaterials > Lipid-Based Structures Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures.
Collapse
Affiliation(s)
- Bin Li
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Xinfu Zhang
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yizhou Dong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, Ohio
- James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
208
|
Ling X, Tu J, Wang J, Shajii A, Kong N, Feng C, Zhang Y, Yu M, Xie T, Bharwani Z, Aljaeid BM, Shi B, Tao W, Farokhzad OC. Glutathione-Responsive Prodrug Nanoparticles for Effective Drug Delivery and Cancer Therapy. ACS NANO 2019; 13:357-370. [PMID: 30485068 PMCID: PMC7049173 DOI: 10.1021/acsnano.8b06400] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Spurred by recent progress in medicinal chemistry, numerous lead compounds have sprung up in the past few years, although the majority are hindered by hydrophobicity, which greatly challenges druggability. In an effort to assess the potential of platinum (Pt) candidates, the nanosizing approach to alter the pharmacology of hydrophobic Pt(IV) prodrugs in discovery and development settings is described. The construction of a self-assembled nanoparticle (NP) platform, composed of amphiphilic lipid-polyethylene glycol (PEG) for effective delivery of Pt(IV) prodrugs capable of resisting thiol-mediated detoxification through a glutathione (GSH)-exhausting effect, offers a promising route to synergistically improving safety and efficacy. After a systematic screening, the optimized NPs (referred to as P6 NPs) exhibited small particle size (99.3 nm), high Pt loading (11.24%), reliable dynamic stability (∼7 days), and rapid redox-triggered release (∼80% in 3 days). Subsequent experiments on cells support the emergence of P6 NPs as a highly effective means of transporting a lethal dose of cargo across cytomembranes through macropinocytosis. Upon reduction by cytoplasmic reductants, particularly GSH, P6 NPs under disintegration released sufficient active Pt(II) metabolites, which covalently bound to target DNA and induced significant apoptosis. The PEGylation endowed P6 NPs with in vivo longevity and tumor specificity, which were essential to successfully inhibiting the growth of cisplatin-sensitive and -resistant xenograft tumors, while effectively alleviating toxic side-effects associated with cisplatin. P6 NPs are, therefore, promising for overcoming the bottleneck in the development of Pt drugs for oncotherapy.
Collapse
Affiliation(s)
- Xiang Ling
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jiasheng Tu
- Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Junqing Wang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Aram Shajii
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Cancer Pharmacology, Holistic Integrative Pharmacy Institutes, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310012, China
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Chan Feng
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ye Zhang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Mikyung Yu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Tian Xie
- Department of Cancer Pharmacology, Holistic Integrative Pharmacy Institutes, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310012, China
- Corresponding Authors:. . .
| | - Zameer Bharwani
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Bader M. Aljaeid
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Corresponding Authors:. . .
| | - Bingyang Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Corresponding Authors:. . .
| | - Omid C. Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Corresponding Authors:. . .
| |
Collapse
|
209
|
Understanding structure-activity relationships of pH-sensitive cationic lipids facilitates the rational identification of promising lipid nanoparticles for delivering siRNAs in vivo. J Control Release 2019; 295:140-152. [PMID: 30610950 DOI: 10.1016/j.jconrel.2019.01.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/21/2018] [Accepted: 01/01/2019] [Indexed: 12/21/2022]
Abstract
Lipid nanoparticles (LNPs) are one of the more promising technologies for efficiently delivering short interfering RNA (siRNA) in vivo. A pH-sensitive cationic lipid that facilitates the targeting of hepatocytes and endosomal escape, strongly influences the availability of siRNA, thus making it a key material for efficient siRNA delivery. A systematic knowledge regarding lipid structure-activity relationships would greatly facilitate the development of sophisticated pH-sensitive cationic lipids for use in siRNA-based therapeutics. The systemic derivatization of a hydrophilic head group and hydrophobic tails of YSK12-C4, a pH-sensitive cationic lipid that was developed in our laboratory, revealed that hydrophilic head significantly affected the apparent pKa of the final product, a key factor in both intrahepatic distribution and endosomal escape. The clogP value of a hydrophilic head group was found to be associated with the apparent pKa of the product. In contrast, the hydrophobic tail structure strongly affected intrahepatic distribution without depending on apparent pKa. A structure-activity relationship study enabled the selection of an adequate combination of a hydrophilic head group and hydrophobic tails and permitted a potent LNP composed of a pH-sensitive cationic lipid CL4H6 (CL4H6-LNPs) to be developed that showed efficient gene silencing activity (50% effective dose: 0.0025 mg/kg), biodegradability and was tolerated. In vivo experiments revealed that the CL4H6-LNPs showed a superior efficiency for endosomal escape, cytosolic release and the RNA-induced silencing for the complex-loading of siRNAs compared to the previously developed LNPs.
Collapse
|
210
|
|
211
|
Kasiewicz LN, Whitehead KA. Lipid nanoparticles silence tumor necrosis factor α to improve wound healing in diabetic mice. Bioeng Transl Med 2019; 4:75-82. [PMID: 30680320 PMCID: PMC6336737 DOI: 10.1002/btm2.10123] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/11/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is a mounting concern in the United States, as are the mortality and morbidity that result from its complications. Of particular concern, diabetes patients frequently suffer from impaired wound healing and resultant nonhealing diabetic foot ulcers. These ulcers overproduce tumor necrosis factor α (TNFα), which reduces wound bed cell migration and proliferation while encouraging apoptosis. Herein, we describe the use of siRNA-loaded lipid nanoparticles (LNPs) as a potential wound treatment to combat an overzealous immune response and facilitate wound closure. LNPs were formulated with an ionizable, degradable lipidoid and siRNA specific for TNFα. Topical application of nanoparticles reduced TNFα mRNA expression in the wound by 40-55% in diabetic and nondiabetic mice. In diabetic mice, this TNFα knockdown accelerated wound healing compared to untreated controls. Together, these results serve as proof-of-concept that RNA interference therapy using LNPs can reduce the severity and duration of chronic diabetic wounds.
Collapse
Affiliation(s)
- Lisa N. Kasiewicz
- Dept. of Chemical EngineeringCarnegie Mellon University5000 Forbes Avenue, PittsburghPA15213
| | - Kathryn A. Whitehead
- Dept. of Chemical EngineeringCarnegie Mellon University5000 Forbes Avenue, PittsburghPA15213
- Dept. of Biomedical EngineeringCarnegie Mellon University5000 Forbes Avenue, PittsburghPA15213
| |
Collapse
|
212
|
Sago CD, Lokugamage MP, Islam FZ, Krupczak BR, Sato M, Dahlman JE. Nanoparticles That Deliver RNA to Bone Marrow Identified by in Vivo Directed Evolution. J Am Chem Soc 2018; 140:17095-17105. [PMID: 30394729 PMCID: PMC6556374 DOI: 10.1021/jacs.8b08976] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone marrow endothelial cells (BMECs) regulate their microenvironment, which includes hematopoietic stem cells. This makes BMECs an important target cell type for siRNA or gene editing (e.g., CRISPR) therapies. However, siRNA and sgRNA have not been delivered to BMECs using systemically administered nanoparticles. Given that in vitro nanoparticle screens have not identified nanoparticles with BMEC tropism, we developed a system to quantify how >100 different nanoparticles deliver siRNA in a single mouse. This is the first barcoding system capable of quantifying functional cytosolic siRNA delivery (where the siRNA drug is active), distinguishing it from in vivo screens that quantify biodistribution (where the siRNA drug went). Combining this approach with bioinformatics, we performed in vivo directed evolution, and identified BM1, a lipid nanoparticle (LNP) that delivers siRNA and sgRNA to BMECs. Interestingly, chemical analysis revealed BMEC tropism was not related to LNP size; tropism changed with the structure of poly(ethylene glycol), as well as the presence of cholesterol. These results suggest that significant changes to vascular targeting can be imparted to a LNP by making simple changes to its chemical composition, rather than using active targeting ligands. BM1 is the first nanoparticle to efficiently deliver siRNA and sgRNA to BMECs in vivo, demonstrating that this functional in vivo screen can identify nanoparticles with novel tropism in vivo. More generally, in vivo screening may help reveal the complex relationship between nanoparticle structure and tropism, thereby helping scientists understand how simple chemical changes control nanoparticle targeting.
Collapse
Affiliation(s)
- Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Fatima Z Islam
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Brandon R Krupczak
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Manaka Sato
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| |
Collapse
|
213
|
Sago CD, Lokugamage MP, Lando GN, Djeddar N, Shah NN, Syed C, Bryksin AV, Dahlman JE. Modifying a Commonly Expressed Endocytic Receptor Retargets Nanoparticles in Vivo. NANO LETTERS 2018; 18:7590-7600. [PMID: 30216729 PMCID: PMC6426696 DOI: 10.1021/acs.nanolett.8b03149] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanoparticles are often targeted to receptors expressed on specific cells, but few receptors are (i) highly expressed on one cell type and (ii) involved in endocytosis. One unexplored alternative is manipulating an endocytic gene expressed on multiple cell types; an ideal gene would inhibit delivery to cell type A more than cell type B, promoting delivery to cell type B. This would require a commonly expressed endocytic gene to alter nanoparticle delivery in a cell type-dependent manner in vivo; whether this can occur is unknown. Based on its microenvironmental regulation, we hypothesized Caveolin 1 (Cav1) would exert cell type-specific effects on nanoparticle delivery. Fluorescence was not sensitive enough to investigate this question, and as a result, we designed a platform named QUANT to study nanoparticle biodistribution. QUANT is 108× more sensitive than fluorescence and can be multiplexed. By measuring how 226 lipid nanoparticles (LNPs) delivered nucleic acids to multiple cell types in vivo in wild-type and Cav1 knockout mice, we found Cav1 altered delivery in a cell-type specific manner. Cav1 knockout did not alter LNP delivery to lung and kidney macrophages but substantially reduced LNP delivery to Kupffer cells, which are liver-resident macrophages. These data suggest caveolin-mediated endocytosis of nanomedicines by macrophages varies with tissue type. These results suggest manipulating receptors expressed on multiple cell types can tune drug delivery.
Collapse
|
214
|
Dunn AW, Kalinichenko VV, Shi D. Highly Efficient In Vivo Targeting of the Pulmonary Endothelium Using Novel Modifications of Polyethylenimine: An Importance of Charge. Adv Healthc Mater 2018; 7:e1800876. [PMID: 30398703 DOI: 10.1002/adhm.201800876] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/26/2018] [Indexed: 01/08/2023]
Abstract
Pulmonary vascular disease encompasses a wide range of serious afflictions with important clinical implications. There is critical need for the development of efficient, nonviral gene therapy delivery systems. Here, a promising avenue to overcome critical issues in efficient cell targeting within the lung via a uniquely designed nanosystem is reported. Polyplexes are created by functionalizing hyperbranched polyethylenimine (PEI) with biological fatty acids and carboxylate-terminated poly(ethylene glycol) (PEG) through a one-pot 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride/N-hydroxysuccinimide reaction. Following intravenous injection, polyplexes show an exceptionally high specificity to the pulmonary microvascular endothelium, allowing for the successful delivery of stabilized enhanced green fluorescent protein (eGFP) expressing messenger ribonucleic acid (mRNA). It is further shown, quantitatively, that positive surface charge is the main mechanism behind such high targeting efficiency for these polyplexes. Live in vivo imaging, flow cytometry of single cell suspensions, and confocal microscopy are used to demonstrate that positive polyplexes are enriched in the lung tissue and disseminated in 85-90% of the alveolar capillary endothelium, whilst being sparse in large vessels. Charge modification, achieved through poly(acrylic acid) or heparin coating, drives a highly significant reduction in both targeting percentage and targeting strength, highlighting the importance of specific surface charge, derived from chemical formulation, for efficient targeting of the pulmonary microvascular endothelium.
Collapse
Affiliation(s)
- Andrew W. Dunn
- The Materials Science and Engineering Program Department of Mechanical and Materials Engineering College of Engineering and Applied Sciences University of Cincinnati Cincinnati OH 45221 USA
| | - Vladimir V. Kalinichenko
- Center for Lung Regenerative Medicine Division of Pulmonary Biology and the Perinatal Institute Cincinnati Children's Hospital Research Foundation Cincinnati OH 45229 USA
| | - Donglu Shi
- The Materials Science and Engineering Program Department of Mechanical and Materials Engineering College of Engineering and Applied Sciences University of Cincinnati Cincinnati OH 45221 USA
| |
Collapse
|
215
|
Kaczmarek JC, Kauffman KJ, Fenton OS, Sadtler K, Patel AK, Heartlein MW, DeRosa F, Anderson DG. Optimization of a Degradable Polymer-Lipid Nanoparticle for Potent Systemic Delivery of mRNA to the Lung Endothelium and Immune Cells. NANO LETTERS 2018; 18:6449-6454. [PMID: 30211557 PMCID: PMC6415675 DOI: 10.1021/acs.nanolett.8b02917] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
mRNA therapeutics hold great potential for treating a variety of diseases through protein-replacement, immunomodulation, and gene editing. However, much like siRNA therapy the majority of progress in mRNA delivery has been confined to the liver. Previously, we demonstrated that poly(β-amino esters), a class of degradable polymers, are capable of systemic mRNA delivery to the lungs in mice when formulated into nanoparticles with poly(ethylene glycol)-lipid conjugates. Using experimental design, a statistical approach to optimization that reduces experimental burden, we demonstrate herein that these degradable polymer-lipid nanoparticles can be optimized in terms of polymer synthesis and nanoparticle formulation to achieve a multiple order-of-magnitude increase in potency. Furthermore, using genetically engineered Cre reporter mice, we demonstrate that mRNA is functionally delivered to both the lung endothelium and pulmonary immune cells, expanding the potential utility of these nanoparticles.
Collapse
Affiliation(s)
- James C. Kaczmarek
- Deparment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA (USA)
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
| | - Kevin J. Kauffman
- Deparment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA (USA)
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
| | - Owen S. Fenton
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA (USA)
| | - Kaitlyn Sadtler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
| | - Asha K. Patel
- Deparment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA (USA)
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham NG7 2RD (UK)
| | | | | | - Daniel G. Anderson
- Deparment of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA (USA)
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA (USA)
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA (USA)
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA (USA)
| |
Collapse
|
216
|
Givens BE, Naguib YW, Geary SM, Devor EJ, Salem AK. Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Therapeutics. AAPS J 2018; 20:108. [PMID: 30306365 PMCID: PMC6398936 DOI: 10.1208/s12248-018-0267-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/18/2018] [Indexed: 12/17/2022] Open
Abstract
The recent progress in harnessing the efficient and precise method of DNA editing provided by CRISPR/Cas9 is one of the most promising major advances in the field of gene therapy. However, the development of safe and optimally efficient delivery systems for CRISPR/Cas9 elements capable of achieving specific targeting of gene therapy to the location of interest without off-target effects is a primary challenge for clinical therapeutics. Nanoparticles (NPs) provide a promising means to meet such challenges. In this review, we present the most recent advances in developing innovative NP-based delivery systems that efficiently deliver CRISPR/Cas9 constructs and maximize their effectiveness.
Collapse
Affiliation(s)
- Brittany E Givens
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Youssef W Naguib
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Sean M Geary
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Eric J Devor
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242, USA
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, 52242, USA.
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa, 52242, USA.
| |
Collapse
|
217
|
Fenton OS, Kauffman KJ, McClellan RL, Kaczmarek JC, Zeng MD, Andresen JL, Rhym LH, Heartlein MW, DeRosa F, Anderson DG. Customizable Lipid Nanoparticle Materials for the Delivery of siRNAs and mRNAs. Angew Chem Int Ed Engl 2018; 57:13582-13586. [PMID: 30112821 PMCID: PMC7548314 DOI: 10.1002/anie.201809056] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Indexed: 12/11/2022]
Abstract
RNAs are a promising class of therapeutics given their ability to regulate protein concentrations at the cellular level. Developing safe and effective strategies to deliver RNAs remains important for realizing their full clinical potential. Here, we develop lipid nanoparticle formulations that can deliver short interfering RNAs (for gene silencing) or messenger RNAs (for gene upregulation). Specifically, we study how the tail length, tail geometry, and linker spacing in diketopiperazine lipid materials influences LNP potency with siRNAs and mRNAs. Eight lipid materials are synthesized, and 16 total formulations are screened for activity in vitro; the lead material is evaluated with mRNA for in vivo use and demonstrates luciferase protein expression in the spleen. In undertaking this approach, not only do we develop synthetic routes to delivery materials, but we also reveal structural criteria that could be useful for developing next-generation delivery materials for RNA therapeutics.
Collapse
Affiliation(s)
- Owen S. Fenton
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Kevin J. Kauffman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Rebecca L. McClellan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - James C. Kaczmarek
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Manhao D. Zeng
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jason L. Andresen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Luke H. Rhym
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | | | - Daniel G. Anderson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge MA 02139
| |
Collapse
|
218
|
Kowalski PS, Bhattacharya C, Afewerki S, Langer R. Smart Biomaterials: Recent Advances and Future Directions. ACS Biomater Sci Eng 2018; 4:3809-3817. [DOI: 10.1021/acsbiomaterials.8b00889] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Piotr S. Kowalski
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Chandrabali Bhattacharya
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Samson Afewerki
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Robert Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
219
|
Fenton OS, Kauffman KJ, McClellan RL, Kaczmarek JC, Zeng MD, Andresen JL, Rhym LH, Heartlein MW, DeRosa F, Anderson DG. Customizable Lipid Nanoparticle Materials for the Delivery of siRNAs and mRNAs. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201809056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Owen S. Fenton
- Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemistry; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Kevin J. Kauffman
- Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Rebecca L. McClellan
- Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - James C. Kaczmarek
- Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Manhao D. Zeng
- Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Jason L. Andresen
- Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemistry; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | - Luke H. Rhym
- Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| | | | | | - Daniel G. Anderson
- Koch Institute for Integrative Cancer Research; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Department of Chemical Engineering; Massachusetts Institute of Technology; Cambridge MA 02139 USA
- Harvard-MIT Division of Health Science and Technology; Massachusetts Institute of Technology; Cambridge MA 02139 USA
| |
Collapse
|
220
|
Facile synthesis of semi-library of low charge density cationic polyesters from poly(alkylene maleate)s for efficient local gene delivery. Biomaterials 2018; 178:559-569. [DOI: 10.1016/j.biomaterials.2018.03.050] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/15/2018] [Accepted: 03/29/2018] [Indexed: 12/20/2022]
|
221
|
Damen M, Groenen AJJ, van Dongen SFM, Nolte RJM, Scholte BJ, Feiters MC. Transfection by cationic gemini lipids and surfactants. MEDCHEMCOMM 2018; 9:1404-1425. [PMID: 30288217 PMCID: PMC6148748 DOI: 10.1039/c8md00249e] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/11/2018] [Indexed: 12/13/2022]
Abstract
Diseases that are linked to defective genes or mutations can in principle be cured by gene therapy, in which damaged or absent genes are either repaired or replaced by new DNA in the nucleus of the cell. Related to this, disorders associated with elevated protein expression levels can be treated by RNA interference via the delivery of siRNA to the cytoplasm of cells. Polynucleotides can be brought into cells by viruses, but this is not without risk for the patient. Alternatively, DNA and RNA can be delivered by transfection, i.e. by non-viral vector systems such as cationic surfactants, which are also referred to as cationic lipids. In this review, recent progress on cationic lipids as transfection vectors will be discussed, with special emphasis on geminis, surfactants with 2 head groups and 2 tails connected by a spacer.
Collapse
Affiliation(s)
- M Damen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - A J J Groenen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - S F M van Dongen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - R J M Nolte
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - B J Scholte
- Departments of Pediatric pulmonology and Cell Biology , Erasmus MC, P. O. Box 2040 , 3000 CA Rotterdam , The Netherlands
| | - M C Feiters
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| |
Collapse
|
222
|
Paunovska K, Gil CJ, Lokugamage MP, Sago CD, Sato M, Lando GN, Gamboa Castro M, Bryksin AV, Dahlman JE. Analyzing 2000 in Vivo Drug Delivery Data Points Reveals Cholesterol Structure Impacts Nanoparticle Delivery. ACS NANO 2018; 12:8341-8349. [PMID: 30016076 PMCID: PMC6115295 DOI: 10.1021/acsnano.8b03640] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Lipid nanoparticles (LNPs) are formulated using unmodified cholesterol. However, cholesterol is naturally esterified and oxidized in vivo, and these cholesterol variants are differentially trafficked in vivo via lipoproteins including LDL and VLDL. We hypothesized that incorporating the same cholesterol variants into LNPs-which can be structurally similar to LDL and VLDL-would alter nanoparticle targeting in vivo. To test this hypothesis, we quantified how >100 LNPs made with six cholesterol variants delivered DNA barcodes to 18 cell types in wild-type, LDLR-/-, and VLDLR-/- mice that were both age-matched and female. By analyzing ∼2000 in vivo drug delivery data points, we found that LNPs formulated with esterified cholesterol delivered nucleic acids more efficiently than LNPs formulated with regular or oxidized cholesterol when compared across all tested cell types in the mouse. We also identified an LNP containing cholesteryl oleate that efficiently delivered siRNA and sgRNA to liver endothelial cells in vivo. Delivery was as-or more-efficient as the same LNP made with unmodified cholesterol. Moreover, delivery to liver endothelial cells was 3 times more efficient than delivery to hepatocytes, distinguishing this oleate LNP from hepatocyte-targeting LNPs. RNA delivery can be improved by rationally selecting cholesterol variants, allowing optimization of nanoparticle targeting.
Collapse
Affiliation(s)
- Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Carmen J Gil
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Manaka Sato
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Gwyn N Lando
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Marielena Gamboa Castro
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Anton V Bryksin
- Petit Institute for Bioengineering and Bioscience , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| |
Collapse
|
223
|
Luu BE, Storey KB. Solving Donor Organ Shortage with Insights from Freeze Tolerance in Nature. Bioessays 2018; 40:e1800092. [DOI: 10.1002/bies.201800092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/02/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Bryan E. Luu
- Institute of Biochemistry and Department of BiologyCarleton University1125 Colonel By DriveOttawaON, K1S 5B6
| | - Kenneth B. Storey
- Institute of Biochemistry and Department of BiologyCarleton University1125 Colonel By DriveOttawaON, K1S 5B6
| |
Collapse
|
224
|
Lokugamage MP, Sago CD, Dahlman JE. Testing thousands of nanoparticles in vivo using DNA barcodes. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 7:1-8. [PMID: 30931416 DOI: 10.1016/j.cobme.2018.08.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nanoparticles improve drug efficacy by delivering drugs to sites of disease. To effectively deliver a drug in vivo, a nanoparticle must overcome physical and physiological hurdles that are not present in cell culture, yet in vitro screens are used to predict nanoparticle delivery in vivo. An ideal nanoparticle discovery pipeline would enable scientists to study thousands of nanoparticles in vivo. Here, we discuss technologies that enable high throughput in vivo screens, focusing on DNA barcoded nanoparticles.
Collapse
Affiliation(s)
- Melissa P Lokugamage
- Address: Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Cory D Sago
- Address: Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - James E Dahlman
- Address: Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| |
Collapse
|
225
|
Shin H, Park SJ, Yim Y, Kim J, Choi C, Won C, Min DH. Recent Advances in RNA Therapeutics and RNA Delivery Systems Based on Nanoparticles. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800065] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Hojeong Shin
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
| | - Se-Jin Park
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
| | - Yeajee Yim
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
| | - Jungho Kim
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
- Institute of Biotherapeutics Convergence Technology; Lemonex Inc.; Seoul 08826 Republic of Korea
| | - Chulwon Choi
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
| | - Cheolhee Won
- Institute of Biotherapeutics Convergence Technology; Lemonex Inc.; Seoul 08826 Republic of Korea
| | - Dal-Hee Min
- Center for RNA Research; Institute for Basic Science; Seoul National University; Seoul 08826 Republic of Korea
- Department of Chemistry; Seoul National University; Seoul 08826 Republic of Korea
- Institute of Biotherapeutics Convergence Technology; Lemonex Inc.; Seoul 08826 Republic of Korea
| |
Collapse
|
226
|
Jang B, Kim B, Kim H, Kwon H, Kim M, Seo Y, Colas M, Jeong H, Jeong EH, Lee K, Lee H. Enzymatic Synthesis of Self-assembled Dicer Substrate RNA Nanostructures for Programmable Gene Silencing. NANO LETTERS 2018; 18:4279-4284. [PMID: 29863365 DOI: 10.1021/acs.nanolett.8b01267] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Enzymatic synthesis of RNA nanostructures is achieved by isothermal rolling circle transcription (RCT). Each arm of RNA nanostructures provides a functional role of Dicer substrate RNA inducing sequence specific RNA interference (RNAi). Three different RNAi sequences (GFP, RFP, and BFP) are incorporated within the three-arm junction RNA nanostructures (Y-RNA). The template and helper DNA strands are designed for the large-scale in vitro synthesis of RNA strands to prepare self-assembled Y-RNA. Interestingly, Dicer processing of Y-RNA is highly influenced by its physical structure and different gene silencing activity is achieved depending on its arm length and overhang. In addition, enzymatic synthesis allows the preparation of various Y-RNA structures using a single DNA template offering on demand regulation of multiple target genes.
Collapse
Affiliation(s)
- Bora Jang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Boyoung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Hyunsook Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Hyokyoung Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Minjeong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Yunmi Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Marion Colas
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
- Faculté de Pharmacie de Paris , Université Paris Descartes , Paris 75006 , France
| | - Hansaem Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Eun Hye Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Kyuri Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences , Ewha Womans University , Seoul 03760 , Republic of Korea
| |
Collapse
|
227
|
Khan OF, Kowalski PS, Doloff JC, Tsosie JK, Bakthavatchalu V, Winn CB, Haupt J, Jamiel M, Langer R, Anderson DG. Endothelial siRNA delivery in nonhuman primates using ionizable low-molecular weight polymeric nanoparticles. SCIENCE ADVANCES 2018; 4:eaar8409. [PMID: 29963629 PMCID: PMC6021147 DOI: 10.1126/sciadv.aar8409] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 05/18/2018] [Indexed: 05/19/2023]
Abstract
Dysfunctional endothelial cells contribute to the pathophysiology of many diseases, including vascular disease, stroke, hypertension, atherosclerosis, organ failure, diabetes, retinopathy, and cancer. Toward the goal of creating a new RNA-based therapy to correct aberrant endothelial cell gene expression in humans, efficient gene silencing in the endothelium of nonhuman primates was achieved by delivering small interfering RNA (siRNA) with 7C1, a low-molecular weight, ionizable polymer that forms nanoparticles. After a single intravenous administration of 1 mg of siRNA per kilogram of animal, 7C1 nanoparticles delivering Tie2 siRNA caused Tie2 mRNA levels to decrease by approximately 80% in the endothelium of the lung. Significant decreases in Tie2 mRNA were also found in the heart, retina, kidney, pancreas, and bone. Blood chemistry and liver function analysis before and after treatment all showed protein and enzyme concentrations within the normal reference ranges. Furthermore, after controlling for siRNA-specific effects, no significant increases in inflammatory cytokine concentrations were found in the serum. Similarly, no gross lesions or significant underlying pathologies were observed after histological examination of nonhuman primate tissues. This study is the first demonstration of endothelial gene silencing in multiple nonhuman primate organs using systemically administered siRNA nanoparticles and highlights the potential of this approach for the treatment of disease in humans.
Collapse
Affiliation(s)
- Omar F. Khan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Piotr S. Kowalski
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Joshua C. Doloff
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jonathan K. Tsosie
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Vasudevan Bakthavatchalu
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 02139
| | - Caroline Bodi Winn
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 02139
| | - Jennifer Haupt
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 02139
| | - Morgan Jamiel
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 02139
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Division of Health Science Technology, Massachusetts Institute of Technology, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G. Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Division of Health Science Technology, Massachusetts Institute of Technology, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Corresponding author.
| |
Collapse
|
228
|
Qiu N, Gao J, Liu Q, Wang J, Shen Y. Enzyme-Responsive Charge-Reversal Polymer-Mediated Effective Gene Therapy for Intraperitoneal Tumors. Biomacromolecules 2018; 19:2308-2319. [DOI: 10.1021/acs.biomac.8b00440] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Nasha Qiu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jinqiang Wang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
229
|
Luo X, Wang W, Dorkin JR, Veiseh O, Chang PH, Abutbul-Ionita I, Danino D, Langer R, Anderson DG, Dong Y. Poly(glycoamidoamine) brush nanomaterials for systemic siRNA delivery in vivo. Biomater Sci 2018; 5:38-40. [PMID: 27921096 DOI: 10.1039/c6bm00683c] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Delivery is the key challenge for siRNA based therapeutics. Here, we report the development of new poly(glycoamidoamine) brush nanomaterials for efficient siRNA delivery. GluN4C10 polymer brush nanoparticles, a lead material, demonstrated significantly improved delivery efficiency for siRNA against factor VII (FVII) in mice compared to poly(glycoamidoamine) brush nanomaterials reported previously.
Collapse
Affiliation(s)
- X Luo
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| | - W Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Department of Anesthesiology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - J R Dorkin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - O Veiseh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Department of Anesthesiology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - P H Chang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Department of Anesthesiology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - I Abutbul-Ionita
- Department of Biotechnology and Food Engineering, Technion Institute of Technology and the Russell Berrie Nanotechnology Institute, Haifa 32000, Israel
| | - D Danino
- Department of Biotechnology and Food Engineering, Technion Institute of Technology and the Russell Berrie Nanotechnology Institute, Haifa 32000, Israel
| | - R Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Department of Anesthesiology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - D G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. and Department of Anesthesiology, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Y Dong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA. and Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA and The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH 43210, USA and The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
230
|
Nikam RR, Gore KR. Journey of siRNA: Clinical Developments and Targeted Delivery. Nucleic Acid Ther 2018; 28:209-224. [PMID: 29584585 DOI: 10.1089/nat.2017.0715] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Since the evolutionary discovery of RNA interference and its utilization for gene knockdown in mammalian cell, a remarkable progress has been achieved in small interfering RNA (siRNA) therapeutics. siRNA is a promising tool, utilized as therapeutic agent against various diseases. Despite its significant potential benefits, safe, efficient, and target oriented delivery of siRNA is one of the major challenges in siRNA therapeutics. This review covers major achievements in clinical trials and targeted delivery of siRNAs using various targeting ligand-receptor pair. Local and systemically administered siRNA drug candidates at various phases in clinical trials are described in this review. This review also provides a deep insight in development of targeted delivery of siRNA. Various targeting ligand-siRNA pair with complexation and conjugation approaches are discussed in this review. This will help to achieve further optimization and development in targeted delivery of siRNAs to achieve higher gene silencing efficiency with lowest siRNA dose availability.
Collapse
Affiliation(s)
| | - Kiran R Gore
- Department of Chemistry, University of Mumbai , Mumbai, India
| |
Collapse
|
231
|
Advances in microfluidics for lipid nanoparticles and extracellular vesicles and applications in drug delivery systems. Adv Drug Deliv Rev 2018; 128:84-100. [PMID: 29567396 DOI: 10.1016/j.addr.2018.03.008] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023]
Abstract
Lipid-based nanobiomaterials as liposomes and lipid nanoparticles (LNPs) are the most widely used nanocarriers for drug delivery systems (DDSs). Extracellular vesicles (EVs) and exosomes are also expected to be applied as DDS nanocarriers. The performance of nanomedicines relies on their components such as lipids, targeting ligands, encapsulated DNA, encapsulated RNA, and drugs. Recently, the importance of the nanocarrier sizes smaller than 100nm is attracting attention as a means to improve nanomedicine performance. Microfluidics and lab-on-a chip technologies make it possible to produce size-controlled LNPs by a simple continuous flow process and to separate EVs from blood samples by using a surface marker, ligand, or electric charge or by making a mass or particle size discrimination. Here, we overview recent advances in microfluidic devices and techniques for liposomes, LNPs, and EVs and their applications for DDSs.
Collapse
|
232
|
Paunovska K, Sago CD, Monaco CM, Hudson WH, Castro MG, Rudoltz TG, Kalathoor S, Vanover DA, Santangelo PJ, Ahmed R, Bryksin AV, Dahlman JE. A Direct Comparison of in Vitro and in Vivo Nucleic Acid Delivery Mediated by Hundreds of Nanoparticles Reveals a Weak Correlation. NANO LETTERS 2018; 18:2148-2157. [PMID: 29489381 PMCID: PMC6054134 DOI: 10.1021/acs.nanolett.8b00432] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Endothelial cells and macrophages play active roles in disease and as a result are important targets for nucleic acid therapies. While thousands of chemically distinct lipid nanoparticles (LNPs) can be synthesized to deliver nucleic acids, studying more than a few LNPs in vivo is challenging. As a result, it is difficult to understand how nanoparticles target these cells in vivo. Using high throughput LNP barcoding, we quantified how well LNPs delivered DNA barcodes to endothelial cells and macrophages in vitro, as well as endothelial cells and macrophages isolated from the lung, heart, and bone marrow in vivo. We focused on two fundamental questions in drug delivery. First, does in vitro LNP delivery predict in vivo LNP delivery? By comparing how 281 LNPs delivered barcodes to endothelial cells and macrophages in vitro and in vivo, we found in vitro delivery did not predict in vivo delivery. Second, does LNP delivery change within the microenvironment of a tissue? We quantified how 85 LNPs delivered barcodes to eight splenic cell populations, and found that cell types derived from myeloid progenitors tended to be targeted by similar LNPs, relative to cell types derived from lymphoid progenitors. These data demonstrate that barcoded LNPs can elucidate fundamental questions about in vivo nanoparticle delivery.
Collapse
Affiliation(s)
- Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Christopher M Monaco
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
- School of Biological Sciences , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - William H Hudson
- Emory Vaccine Center and Department of Microbiology and Immunology , Emory University School of Medicine , Atlanta , Georgia 30317 , United States
| | - Marielena Gamboa Castro
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Tobi G Rudoltz
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Sujay Kalathoor
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Daryll A Vanover
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology , Emory University School of Medicine , Atlanta , Georgia 30317 , United States
| | - Anton V Bryksin
- Parker H. Petit Institute for Bioengineering and Bioscience , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| |
Collapse
|
233
|
Miller JB, Kos P, Tieu V, Zhou K, Siegwart DJ. Development of Cationic Quaternary Ammonium Sulfonamide Amino Lipids for Nucleic Acid Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:2302-2311. [PMID: 29286232 DOI: 10.1021/acsami.7b15982] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Lipid nanoparticles (LNPs) currently comprise the most effective carrier class for the delivery of small RNAs. Among lipid carriers, charge-unbalanced lipids are relatively unexplored synthetically. Herein, we developed and evaluated a novel collection of compounds for small interfering RNA (siRNA) delivery, termed cationic quaternary ammonium sulfonamide amino lipids (CSALs). The formulated CSAL LNPs containing cholesterol, 1,2-distearoyl-sn-glycero-3-phosphocholine, and lipid poly(ethylene glycol) exhibited biophysical property trends directly related to the CSAL chemical structure. Lead CSAL LNPs were identified using an siRNA delivery screen. Further chemical synthesis using a rational structure-guided design showed that the head group structure could alter the pKa and other physical properties that modulated delivery efficacy. Shorter-chained dimethylamino head groups, acetate side chains, and higher tail carbon numbers were favorable for delivery. This led to a further study of A3-OAc-C2Me LNPs, which enabled in vivo delivery to normal mouse lungs and subcutaneous and orthotopic lung tumors. Incorporation of CSALs into liver-targeting formulations shifted the in vivo delivery of these carriers to the lungs. This study highlights the importance of the cationic lipid structure in LNPs and provides further design guidelines for nucleic acid carriers.
Collapse
Affiliation(s)
- Jason B Miller
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Petra Kos
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Victor Tieu
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Kejin Zhou
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Daniel J Siegwart
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| |
Collapse
|
234
|
McLoughlin NM, Mueller C, Grossmann TN. The Therapeutic Potential of PTEN Modulation: Targeting Strategies from Gene to Protein. Cell Chem Biol 2018; 25:19-29. [DOI: 10.1016/j.chembiol.2017.10.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/05/2017] [Accepted: 10/23/2017] [Indexed: 01/04/2023]
|
235
|
Sager HB, Dutta P, Dahlman JE, Hulsmans M, Courties G, Sun Y, Heidt T, Vinegoni C, Borodovsky A, Fitzgerald K, Wojtkiewicz GR, Iwamoto Y, Tricot B, Khan OF, Kauffman KJ, Xing Y, Shaw TE, Libby P, Langer R, Weissleder R, Swirski FK, Anderson DG, Nahrendorf M. RNAi targeting multiple cell adhesion molecules reduces immune cell recruitment and vascular inflammation after myocardial infarction. Sci Transl Med 2017; 8:342ra80. [PMID: 27280687 DOI: 10.1126/scitranslmed.aaf1435] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 05/17/2016] [Indexed: 12/22/2022]
Abstract
Myocardial infarction (MI) leads to a systemic surge of vascular inflammation in mice and humans, resulting in secondary ischemic complications and high mortality. We show that, in ApoE(-/-) mice with coronary ligation, increased sympathetic tone up-regulates not only hematopoietic leukocyte production but also plaque endothelial expression of adhesion molecules. To counteract the resulting arterial leukocyte recruitment, we developed nanoparticle-based RNA interference (RNAi) that effectively silences five key adhesion molecules. Simultaneously encapsulating small interfering RNA (siRNA)-targeting intercellular cell adhesion molecules 1 and 2 (Icam1 and Icam2), vascular cell adhesion molecule 1 (Vcam1), and E- and P-selectins (Sele and Selp) into polymeric endothelial-avid nanoparticles reduced post-MI neutrophil and monocyte recruitment into atherosclerotic lesions and decreased matrix-degrading plaque protease activity. Five-gene combination RNAi also curtailed leukocyte recruitment to ischemic myocardium. Therefore, targeted multigene silencing may prevent complications after acute MI.
Collapse
Affiliation(s)
- Hendrik B Sager
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Partha Dutta
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - James E Dahlman
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA
| | - Maarten Hulsmans
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Gabriel Courties
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yuan Sun
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Timo Heidt
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | - Gregory R Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Benoit Tricot
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Omar F Khan
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Kevin J Kauffman
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Yiping Xing
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Taylor E Shaw
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert Langer
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA.,Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Daniel G Anderson
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA.,Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.,Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
236
|
Yin H, Song CQ, Suresh S, Wu Q, Walsh S, Rhym LH, Mintzer E, Bolukbasi MF, Zhu LJ, Kauffman K, Mou H, Oberholzer A, Ding J, Kwan SY, Bogorad RL, Zatsepin T, Koteliansky V, Wolfe SA, Xue W, Langer R, Anderson DG. Structure-guided chemical modification of guide RNA enables potent non-viral in vivo genome editing. Nat Biotechnol 2017; 35:1179-1187. [PMID: 29131148 PMCID: PMC5901668 DOI: 10.1038/nbt.4005] [Citation(s) in RCA: 337] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 10/09/2017] [Indexed: 12/12/2022]
Abstract
Efficient genome editing with Cas9-sgRNA in vivo has required the use of viral delivery systems, which have limitations for clinical applications. Translational efforts to develop other RNA therapeutics have shown that judicious chemical modification of RNAs can improve therapeutic efficacy by reducing susceptibility to nuclease degradation. Guided by the structure of the Cas9-sgRNA complex, we identify regions of sgRNA that can be modified while maintaining or enhancing genome-editing activity, and we develop an optimal set of chemical modifications for in vivo applications. Using lipid nanoparticle formulations of these enhanced sgRNAs (e-sgRNA) and mRNA encoding Cas9, we show that a single intravenous injection into mice induces >80% editing of Pcsk9 in the liver. Serum Pcsk9 is reduced to undetectable levels, and cholesterol levels are significantly lowered about 35% to 40% in animals. This strategy may enable non-viral, Cas9-based genome editing in the liver in clinical settings.
Collapse
Affiliation(s)
- Hao Yin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Chun-Qing Song
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sneha Suresh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Qiongqiong Wu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Stephen Walsh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Luke Hyunsik Rhym
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Esther Mintzer
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Mehmet Fatih Bolukbasi
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kevin Kauffman
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Haiwei Mou
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Alicia Oberholzer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Junmei Ding
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Suet-Yan Kwan
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Roman L Bogorad
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Timofei Zatsepin
- Center of Translational Biomedicine, Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Victor Koteliansky
- Center of Translational Biomedicine, Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts, USA
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts, USA
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
237
|
Kauffman KJ, Oberli MA, Dorkin JR, Hurtado JE, Kaczmarek JC, Bhadani S, Wyckoff J, Langer R, Jaklenec A, Anderson DG. Rapid, Single-Cell Analysis and Discovery of Vectored mRNA Transfection In Vivo with a loxP-Flanked tdTomato Reporter Mouse. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 10:55-63. [PMID: 29499956 PMCID: PMC5734870 DOI: 10.1016/j.omtn.2017.11.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/09/2017] [Accepted: 11/15/2017] [Indexed: 01/01/2023]
Abstract
mRNA therapeutics hold promise for the treatment of diseases requiring intracellular protein expression and for use in genome editing systems, but mRNA must transfect the desired tissue and cell type to be efficacious. Nanoparticle vectors that deliver the mRNA are often evaluated using mRNA encoding for reporter genes such as firefly luciferase (FLuc); however, single-cell resolution of mRNA expression cannot generally be achieved with FLuc, and, thus, the transfected cell populations cannot be determined without additional steps or experiments. To more rapidly identify which types of cells an mRNA formulation transfects in vivo, we describe a Cre recombinase (Cre)-based system that permanently expresses fluorescent tdTomato protein in transfected cells of genetically modified mice. Following in vivo application of vectored Cre mRNA, it is possible to visualize successfully transfected cells via Cre-mediated tdTomato expression in bulk tissues and with single-cell resolution. Using this system, we identify previously unknown transfected cell types of an existing mRNA delivery vehicle in vivo and also develop a new mRNA formulation capable of transfecting lung endothelial cells. Importantly, the same formulations with mRNA encoding for fluorescent protein delivered to wild-type mice did not produce sufficient signal for any visualization in vivo, demonstrating the significantly improved sensitivity of our Cre-based system. We believe that the system described here may facilitate the identification and characterization of mRNA delivery vectors to new tissues and cell types.
Collapse
Affiliation(s)
- Kevin J Kauffman
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Matthias A Oberli
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - J Robert Dorkin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Juan E Hurtado
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - James C Kaczmarek
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shivani Bhadani
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeff Wyckoff
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139.
| |
Collapse
|
238
|
Panek WK, Khan OF, Yu D, Lesniak MS. Multiplexed nanomedicine for brain tumors: nanosized Hercules to tame our Lernaean Hydra inside? Nanomedicine (Lond) 2017; 12:2435-2439. [PMID: 28971724 DOI: 10.2217/nnm-2017-0260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Wojciech K Panek
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Omar F Khan
- David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Institute for Medical Engineering & Science, Harvard MIT Division of Health Science & Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dou Yu
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
239
|
Mechanistic understanding of in vivo protein corona formation on polymeric nanoparticles and impact on pharmacokinetics. Nat Commun 2017; 8:777. [PMID: 28974673 PMCID: PMC5626760 DOI: 10.1038/s41467-017-00600-w] [Citation(s) in RCA: 448] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/11/2017] [Indexed: 01/27/2023] Open
Abstract
In vitro incubation of nanomaterials with plasma offer insights on biological interactions, but cannot fully explain the in vivo fate of nanomaterials. Here, we use a library of polymer nanoparticles to show how physicochemical characteristics influence blood circulation and early distribution. For particles with different diameters, surface hydrophilicity appears to mediate early clearance. Densities above a critical value of approximately 20 poly(ethylene glycol) chains (MW 5 kDa) per 100 nm2 prolong circulation times, irrespective of size. In knockout mice, clearance mechanisms are identified for nanoparticles with low and high steric protection. Studies in animals deficient in the C3 protein showed that complement activation could not explain differences in the clearance of nanoparticles. In nanoparticles with low poly(ethylene glycol) coverage, adsorption of apolipoproteins can prolong circulation times. In parallel, the low-density-lipoprotein receptor plays a predominant role in the clearance of nanoparticles, irrespective of poly(ethylene glycol) density. These results further our understanding of nanopharmacology. Understanding the interaction between nanoparticles and biomolecules is crucial for improving current drug-delivery systems. Here, the authors shed light on the essential role of the surface and other physicochemical properties of a library of nanoparticles on their in vivo pharmacokinetics.
Collapse
|
240
|
Riber CF, Andersen AHF, Rolskov LA, Zuwala K, Gajda P, Løvschall KB, Dagnæs-Hansen F, Banda DH, Pietschmann T, Tolstrup M, Zelikin AN. Synthetic Polymer with a Structure-Driven Hepatic Deposition and Curative Pharmacological Activity in Hepatic Cells. ACS Macro Lett 2017; 6:935-940. [PMID: 35650894 DOI: 10.1021/acsmacrolett.7b00471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Synthetic polymers make strong contributions as tools for delivery of biological drugs and chemotherapeutics. The most praised characteristic of polymers in these applications is complete lack of pharmacological function such as to minimize the side effects within the human body. In contrast, synthetic polymers with curative pharmacological activity are truly rare. Moreover, such activity is typically nonspecific rather than structure-defined. In this work, we present the discovery of poly(ethylacrylic acid) (PEAA) as a polymer with a suit of structure-defined, unexpected, pharmacological, and pharmacokinetic properties not observed in close structural analogues. Specifically, PEAA reveals capacity to bind to albumin with ensuing natural hepatic deposition in vivo and exhibits concurrent inhibitory activity against the hepatitis C virus and inflammation in hepatic cells. Our findings provide a view on synthetic polymers as curative, functional agents and present PEAA as a unique biomedical tool with applications related to health of the human liver.
Collapse
Affiliation(s)
- Camilla Frich Riber
- Department
of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Anna Halling Folkmar Andersen
- Department
of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
- Department
of Infectious Diseases, Aarhus University Hospital, Aarhus 8200, Denmark
- Department
of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | - Lærke Anegaard Rolskov
- Department
of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | - Kaja Zuwala
- Department
of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
- Department
of Infectious Diseases, Aarhus University Hospital, Aarhus 8200, Denmark
- Department
of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | - Paulina Gajda
- Department
of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
- Department
of Infectious Diseases, Aarhus University Hospital, Aarhus 8200, Denmark
- Department
of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | - Kaja Borup Løvschall
- Department
of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| | | | - Dominic H. Banda
- Institute
of Experimental Virology, TWINCORE Centre for Experimental and Clinical
Infection Research, Medical School Hannover/Helmholtz Centre for Infection Research, Hannover, Germany
| | - Thomas Pietschmann
- Institute
of Experimental Virology, TWINCORE Centre for Experimental and Clinical
Infection Research, Medical School Hannover/Helmholtz Centre for Infection Research, Hannover, Germany
| | - Martin Tolstrup
- Department
of Infectious Diseases, Aarhus University Hospital, Aarhus 8200, Denmark
- Department
of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | - Alexander N. Zelikin
- Department
of Chemistry and iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus 8000, Denmark
| |
Collapse
|
241
|
Patel S, Ashwanikumar N, Robinson E, DuRoss A, Sun C, Murphy-Benenato KE, Mihai C, Almarsson Ö, Sahay G. Boosting Intracellular Delivery of Lipid Nanoparticle-Encapsulated mRNA. NANO LETTERS 2017; 17:5711-5718. [PMID: 28836442 PMCID: PMC5623340 DOI: 10.1021/acs.nanolett.7b02664] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Intracellular delivery of mRNA holds great potential for vaccine1-3 and therapeutic4 discovery and development. Despite increasing recognition of the utility of lipid-based nanoparticles (LNPs) for intracellular delivery of mRNA, particle engineering is hindered by insufficient understanding of endosomal escape, which is believed to be a main limiter of cytosolic availability and activity of the nucleic acid inside the cell. Using a series of CRISPR-based genetic perturbations of the lysosomal pathway, we have identified that late endosome/lysosome (LE/Ly) formation is essential for functional delivery of exogenously presented mRNA. Lysosomes provide a spatiotemporal hub to orchestrate mTOR signaling and are known to control cell proliferation, nutrient sensing, ribosomal biogenesis, and mRNA translation. Through modulation of the mTOR pathway we were able to enhance or inhibit LNP-mediated mRNA delivery. To further boost intracellular delivery of mRNA, we screened 212 bioactive lipid-like molecules that are either enriched in vesicular compartments or modulate cell signaling. Surprisingly, we have discovered that leukotriene-antagonists, clinically approved for treatment of asthma and other lung diseases, enhance intracellular mRNA delivery in vitro (over 3-fold, p < 0.005) and in vivo (over 2-fold, p < 0.005). Understanding LNP-mediated intracellular delivery will inspire the next generation of RNA therapeutics that have high potency and limited toxicity.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - N Ashwanikumar
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Emily Robinson
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Allison DuRoss
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Conroy Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
- Department of Radiation Medicine, School of Medicine, 3181 S.W. Sam Jackson Park Road, Oregon Health Science University, Portland, OR, 97239
| | | | - Cosmin Mihai
- Moderna Therapeutics, 200 Technology Square, Cambridge, MA, 02139
| | - Örn Almarsson
- Moderna Therapeutics, 200 Technology Square, Cambridge, MA, 02139
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
- Department of Biomedical Engineering, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon Health Science University, Portland, OR, 97201
| |
Collapse
|
242
|
Chen D, Ganesh S, Wang W, Amiji M. Plasma protein adsorption and biological identity of systemically administered nanoparticles. Nanomedicine (Lond) 2017; 12:2113-2135. [DOI: 10.2217/nnm-2017-0178] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although a variety of nanoparticles (NPs) have been used for drug delivery applications, their surfaces are immediately covered by plasma protein corona upon systemic administration. As a result, the adsorbed proteins create a unique biological identity of the NPs that lead to unpredictable performance. The protein corona on NPs could also impede active targeting, induce off-target effects, trigger particle clearance and even provoke toxicity. This article reviews the fundamentals of NP–plasma protein interaction, the consequences of the interactions, and provides insights into the correlations of protein corona with biodistribution and cellular delivery. We hope that this review will trigger additional questions and possible solutions that lead to more favorable developments in NP-based targeted delivery systems.
Collapse
Affiliation(s)
- Dongyu Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| | - Shanthi Ganesh
- Department of Pre-Clinical Oncology, Dicerna Pharmaceuticals, Inc., Cambridge, MA 02140, USA
| | - Weimin Wang
- Department of Chemistry and Formulation, Dicerna Pharmaceuticals, Inc., Cambridge, MA 02140, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
243
|
Zhang X, Li B, Luo X, Zhao W, Jiang J, Zhang C, Gao M, Chen X, Dong Y. Biodegradable Amino-Ester Nanomaterials for Cas9 mRNA Delivery in Vitro and in Vivo. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25481-25487. [PMID: 28685575 PMCID: PMC5896755 DOI: 10.1021/acsami.7b08163] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Efficient and safe delivery of the CRISPR/Cas system is one of the key challenges for genome-editing applications in humans. Herein, we designed and synthesized a series of biodegradable lipidlike compounds containing ester groups for the delivery of mRNA-encoding Cas9. Two lead materials, termed N-methyl-1,3-propanediamine (MPA)-A and MPA-Ab, showed a tunable rate of biodegradation. MPA-A with linear ester chains was degraded dramatically faster than MPA-Ab with branched ester chains in the presence of esterase or in wild-type mice. Most importantly, MPA-A and MPA-Ab demonstrated efficient delivery of Cas9 mRNA both in vitro and in vivo. Consequently, these biodegradable lipidlike nanomaterials merit further development as genome-editing delivery tools for biological and therapeutic applications.
Collapse
Affiliation(s)
- Xinfu Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Bin Li
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Xiao Luo
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Weiyu Zhao
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Justin Jiang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chengxiang Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Min Gao
- LCI Characterization Facility, Liquid Crystal Institute, Kent State University, Kent, Ohio 44242, United States
| | - Xiaofang Chen
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- The Center for Clinical and Translational Science, The Ohio State University, Columbus, Ohio 43210, United States
- The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
244
|
Xiong H, Zuo H, Yan Y, Occhialini G, Zhou K, Wan Y, Siegwart DJ. High-Contrast Fluorescence Detection of Metastatic Breast Cancer Including Bone and Liver Micrometastases via Size-Controlled pH-Activatable Water-Soluble Probes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1700131. [PMID: 28563903 DOI: 10.1002/adma.201700131] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/19/2017] [Indexed: 06/07/2023]
Abstract
Breast cancer metastasis is the major cause of cancer death in women worldwide. Early detection would save many lives, but current fluorescence imaging probes are limited in their detection ability, particularly of bone and liver micrometastases. Herein, probes that are capable of imaging tiny (<1 mm) micrometastases in the liver, lung, pancreas, kidneys, and bone, that have disseminated from the primary site, are reported. The influence of the poly(ethylene glycol) (PEG) chain length on the performance of water-soluble, pH-responsive, near-infrared 4,4'-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY) probes is systematically investigated to demonstrate that PEG tuning can provide control over micrometastasis tracking with high tumor-to-background contrast (up to 12/1). Optimized probes can effectively visualize tumor boundaries and successfully detect micrometastases with diameters <1 mm. The bone-metastasis-targeting ability of these probes is further enhanced by covalent functionalization with bisphosphonate. This improved detection of both bone and liver micrometastases (<2 mm) with excellent tumor-to-normal contrast (5.2/1). A versatile method is thus introduced to directly synthesize modular water-soluble probes with broad potential utility. Through a single intravenous injection, these materials can image micrometastases in multiple organs with spatiotemporal resolution. They thus hold promise for metastasis diagnosis, image-guided surgery, and theranostic PEGylated drug therapies.
Collapse
Affiliation(s)
- Hu Xiong
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hao Zuo
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yunfeng Yan
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Gino Occhialini
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kejin Zhou
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yihong Wan
- Department of Pharmacology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel J Siegwart
- Simmons Comprehensive Cancer Center, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
245
|
Hashiba K, Sato Y, Harashima H. pH-labile PEGylation of siRNA-loaded lipid nanoparticle improves active targeting and gene silencing activity in hepatocytes. J Control Release 2017; 262:239-246. [PMID: 28774839 DOI: 10.1016/j.jconrel.2017.07.046] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/03/2017] [Accepted: 07/31/2017] [Indexed: 11/17/2022]
Abstract
Lipid nanoparticles (LNPs) are one of the promising technologies for the in vivo delivery of short interfering RNA (siRNA). Modifying LNPs with polyethyleneglycol (PEG) is widely used to inhibit non-specific interactions with serum components in the blood stream, and is a useful strategy for maximizing the efficiency of active targeting. However, it is a widely accepted fact that PEGylation of the LNP surface strongly inhibits fusion between LNPs and endosomal membranes, resulting in poor cytosolic siRNA delivery, a process that is referred to as the 'PEG-dilemma'. In the present study, in an attempt to overcome this problem, siRNA-loaded LNPs were modified with PEG through maleic anhydride, a pH-labile linkage. The in vitro, suppression of cationic charge, stealth function at physiological pH up to 1h and the rapid desorption of PEG and restoration of fusogenic activity under slightly acidic conditions (within only 2min) were achieved by PEG modification of the LNPs through maleic anhydride. In vivo, PEG modification through maleic anhydride resulted in a dramatic improvement in the targeting capability of the active targeting of ligand (N-acetyl-d-galactosamine)-modified LNPs to hepatocytes, with an approximately 14-fold increase in gene silencing activity in factor 7 model mice. Taken together, the maleic anhydride-mediated pH-labile PEGylation of the active targeting LNPs is a useful strategy for achieving the specific and efficient delivery of siRNAs in vivo.
Collapse
Affiliation(s)
- Kazuki Hashiba
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo 060-0812, Japan
| | - Yusuke Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
246
|
Yu D, Khan OF, Suvà ML, Dong B, Panek WK, Xiao T, Wu M, Han Y, Ahmed AU, Balyasnikova IV, Zhang HF, Sun C, Langer R, Anderson DG, Lesniak MS. Multiplexed RNAi therapy against brain tumor-initiating cells via lipopolymeric nanoparticle infusion delays glioblastoma progression. Proc Natl Acad Sci U S A 2017; 114:E6147-E6156. [PMID: 28696296 PMCID: PMC5544292 DOI: 10.1073/pnas.1701911114] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Brain tumor-initiating cells (BTICs) have been identified as key contributors to therapy resistance, recurrence, and progression of diffuse gliomas, particularly glioblastoma (GBM). BTICs are elusive therapeutic targets that reside across the blood-brain barrier, underscoring the urgent need to develop novel therapeutic strategies. Additionally, intratumoral heterogeneity and adaptations to therapeutic pressure by BTICs impede the discovery of effective anti-BTIC therapies and limit the efficacy of individual gene targeting. Recent discoveries in the genetic and epigenetic determinants of BTIC tumorigenesis offer novel opportunities for RNAi-mediated targeting of BTICs. Here we show that BTIC growth arrest in vitro and in vivo is accomplished via concurrent siRNA knockdown of four transcription factors (SOX2, OLIG2, SALL2, and POU3F2) that drive the proneural BTIC phenotype delivered by multiplexed siRNA encapsulation in the lipopolymeric nanoparticle 7C1. Importantly, we demonstrate that 7C1 nano-encapsulation of multiplexed RNAi is a viable BTIC-targeting strategy when delivered directly in vivo in an established mouse brain tumor. Therapeutic potential was most evident via a convection-enhanced delivery method, which shows significant extension of median survival in two patient-derived BTIC xenograft mouse models of GBM. Our study suggests that there is potential advantage in multiplexed targeting strategies for BTICs and establishes a flexible nonviral gene therapy platform with the capacity to channel multiplexed RNAi schemes to address the challenges posed by tumor heterogeneity.
Collapse
Affiliation(s)
- Dou Yu
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Omar F Khan
- Department of Chemical Engineering, Institute for Medical Engineering and Science, Harvard MIT Division of Health Science and Technology, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Mario L Suvà
- Broad Institute of Harvard and MIT, Cambridge, MA 02142
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Biqin Dong
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208
- Department of Mechanical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208
| | - Wojciech K Panek
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Ting Xiao
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Meijing Wu
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Yu Han
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Atique U Ahmed
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Hao F Zhang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208
- Department of Mechanical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208
| | - Cheng Sun
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208
| | - Robert Langer
- Department of Chemical Engineering, Institute for Medical Engineering and Science, Harvard MIT Division of Health Science and Technology, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Daniel G Anderson
- Department of Chemical Engineering, Institute for Medical Engineering and Science, Harvard MIT Division of Health Science and Technology, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Maciej S Lesniak
- Department of Neurological Surgery, Brain Tumor Research Institute, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611;
| |
Collapse
|
247
|
Lipophilic siRNA targets albumin in situ and promotes bioavailability, tumor penetration, and carrier-free gene silencing. Proc Natl Acad Sci U S A 2017; 114:E6490-E6497. [PMID: 28739942 DOI: 10.1073/pnas.1621240114] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Clinical translation of therapies based on small interfering RNA (siRNA) is hampered by siRNA's comprehensively poor pharmacokinetic properties, which necessitate molecule modifications and complex delivery strategies. We sought an alternative approach to commonly used nanoparticle carriers by leveraging the long-lived endogenous serum protein albumin as an siRNA carrier. We synthesized siRNA conjugated to a diacyl lipid moiety (siRNA-L2), which rapidly binds albumin in situ. siRNA-L2, in comparison with unmodified siRNA, exhibited a 5.7-fold increase in circulation half-life, an 8.6-fold increase in bioavailability, and reduced renal accumulation. Benchmarked against leading commercial siRNA nanocarrier in vivo jetPEI, siRNA-L2 achieved 19-fold greater tumor accumulation and 46-fold increase in per-tumor-cell uptake in a mouse orthotopic model of human triple-negative breast cancer. siRNA-L2 penetrated tumor tissue rapidly and homogeneously; 30 min after i.v. injection, siRNA-L2 achieved uptake in 99% of tumor cells, compared with 60% for jetPEI. Remarkably, siRNA-L2 achieved a tumor:liver accumulation ratio >40:1 vs. <3:1 for jetPEI. The improved pharmacokinetic properties of siRNA-L2 facilitated significant tumor gene silencing for 7 d after two i.v. doses. Proof-of-concept was extended to a patient-derived xenograft model, in which jetPEI tumor accumulation was reduced fourfold relative to the same formulation in the orthotopic model. The siRNA-L2 tumor accumulation diminished only twofold, suggesting that the superior tumor distribution of the conjugate over nanoparticles will be accentuated in clinical situations. These data reveal the immense promise of in situ albumin targeting for development of translational, carrier-free RNAi-based cancer therapies.
Collapse
|
248
|
Gossart JB, Pascal E, Meyer F, Heuillard E, Gonçalves M, Gossé F, Robinet E, Frisch B, Seguin C, Zuber G. Performance of Pyridylthiourea-Polyethylenimine Polyplex for siRNA-Mediated Liver Cancer Therapy in Cell Monolayer, Spheroid, and Tumor Xenograft Models. GLOBAL CHALLENGES (HOBOKEN, NJ) 2017; 1:1700013. [PMID: 31565271 PMCID: PMC6607116 DOI: 10.1002/gch2.201700013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/14/2017] [Indexed: 06/10/2023]
Abstract
Medical application of siRNAs relies on methods for delivering nucleic acids into the cytosol. Synthetic carriers, which assemble with nucleic acids into delivery systems, show promises for cancer therapy but efficiency remains to be improved. In here, the effectiveness of pyridylthiourea-polyethylenimine (πPEI), a siRNA carrier that favors both polyplex disassembly and endosome rupture upon sensing the acidic endosomal environment, in 3 experimental models of hepatocellular cancer is tested. The πPEI-assisted delivery of a siRNA targeting the polo-like kinase 1 into Huh-7 monolayer produces a 90% cell death via a demonstrated RNA interference mechanism. Incubation of polyplex with Huh-7 spheroids leads to siRNA delivery into the superficial first cell layer and a 60% reduction in spheroid growth compared to untreated controls. Administration of polyplexes into mice bearing subcutaneous implanted Huh-7Luc tumors results in a reduced tumor progression, similar to the one observed in the spheroid model. Altogether, these results support the in vivo use of synthetic and dedicated polymers for increasing siRNA-mediated gene knockdown, and their clinical promise in cancer therapeutics.
Collapse
Affiliation(s)
- Jean Baptiste Gossart
- Université de Strasbourg‐CNRS CAMB UMR 7199Faculté de Pharmacie74 route du Rhin67400IllkirchFrance
- Université de Strasbourg‐INSERMUMRS 1121 Biomaterials and Bioengineering, FTMS11 rue Humann67000StrasbourgFrance
| | - Etienne Pascal
- Université de Strasbourg‐CNRS CAMB UMR 7199Faculté de Pharmacie74 route du Rhin67400IllkirchFrance
| | - Florent Meyer
- Université de Strasbourg‐INSERMUMRS 1121 Biomaterials and Bioengineering, FTMS11 rue Humann67000StrasbourgFrance
| | - Emilie Heuillard
- Institut Hospitalo‐Universitaire de Strasbourg1 place de l'Hôpital67000StrasbourgFrance
| | - Mathieu Gonçalves
- Institut Hospitalo‐Universitaire de Strasbourg1 place de l'Hôpital67000StrasbourgFrance
| | - Francine Gossé
- Institut Hospitalo‐Universitaire de Strasbourg1 place de l'Hôpital67000StrasbourgFrance
- Inserm U11103 rue Koeberlé67000StrasbourgFrance
| | - Eric Robinet
- Institut Hospitalo‐Universitaire de Strasbourg1 place de l'Hôpital67000StrasbourgFrance
- Inserm U11103 rue Koeberlé67000StrasbourgFrance
| | - Benoît Frisch
- Université de Strasbourg‐CNRS CAMB UMR 7199Faculté de Pharmacie74 route du Rhin67400IllkirchFrance
| | - Cendrine Seguin
- Université de Strasbourg‐CNRS CAMB UMR 7199Faculté de Pharmacie74 route du Rhin67400IllkirchFrance
| | - Guy Zuber
- Université de Strasbourg‐CNRS, UMR 7242Boulevard Sebastien Brant67400IllkirchFrance
| |
Collapse
|
249
|
In vitro 3D model and miRNA drug delivery to target calcific aortic valve disease. Clin Sci (Lond) 2017; 131:181-195. [PMID: 28057890 DOI: 10.1042/cs20160378] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 10/27/2016] [Accepted: 11/14/2016] [Indexed: 12/20/2022]
Abstract
Calcific aortic valve disease (CAVD) is the most prevalent valvular heart disease in the Western population, claiming 17000 deaths per year in the United States and affecting 25% of people older than 65 years of age. Contrary to traditional belief, CAVD is not a passive, degenerative disease but rather a dynamic disease, where initial cellular changes in the valve leaflets progress into fibrotic lesions that induce valve thickening and calcification. Advanced thickening and calcification impair valve function and lead to aortic stenosis (AS). Without intervention, progressive ventricular hypertrophy ensues, which ultimately results in heart failure and death. Currently, aortic valve replacement (AVR), surgical or transcatheter, is the only effective therapy to treat CAVD. However, these costly interventions are often delayed until the late stages of the disease. Nonetheless, 275000 are performed per year worldwide, and this is expected to triple by 2050. Given the current landscape, next-generation therapies for CAVD are needed to improve patient outcome and quality of life. Here, we first provide a background on the aortic valve (AV) and the pathobiology of CAVD as well as highlight current directions and future outlook on the development of functional 3D models of CAVD in vitro We then consider an often-overlooked aspect contributing to CAVD: miRNA (mis)regulation. Therapeutics could potentially normalize miRNA levels in the early stages of the disease and may slow its progression or even reverse calcification. We close with a discussion of strategies that would enable the use of miRNA as a therapeutic for CAVD. This focuses on an overview of controlled delivery technologies for nucleic acid therapeutics to the valve or other target tissues.
Collapse
|
250
|
Acharya R, Saha S, Ray S, Hazra S, Mitra MK, Chakraborty J. siRNA-nanoparticle conjugate in gene silencing: A future cure to deadly diseases? MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:1378-1400. [DOI: 10.1016/j.msec.2017.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 01/17/2017] [Accepted: 03/01/2017] [Indexed: 02/08/2023]
|