1
|
Park W, Choi J, Hwang J, Kim S, Kim Y, Shim MK, Park W, Yu S, Jung S, Yang Y, Kweon DH. Apolipoprotein Fusion Enables Spontaneous Functionalization of mRNA Lipid Nanoparticles with Antibody for Targeted Cancer Therapy. ACS NANO 2025; 19:6412-6425. [PMID: 39908463 PMCID: PMC11841042 DOI: 10.1021/acsnano.4c16562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
The mRNA-lipid nanoparticles (mRNA@LNPs) offer a novel opportunity to treat targets previously considered undruggable. Although antibody conjugation is crucial for enhancing the specificity, delivery efficiency, and minimizing the toxicity of mRNA therapeutics, current chemical conjugation methods are complex and produce heterogeneous particles with misoriented antibodies. In this work, we introduce a chemical-free approach to functionalize mRNA@LNPs with antibodies, mimicking protein corona formation for targeted mRNA delivery. By fusing apolipoprotein to the Fc domain of a targeting antibody, we enabled the antibody to spontaneously display on the surface of mRNA@LNPs without altering the existing LNP process or employing complex chemical conjugation techniques. We demonstrated precise protein expression using trastuzumab-bound mRNA@LNPs, facilitating specific mRNA expression in HER2-positive cancer cells. mRNA was efficiently delivered to the tumor site after intravenous administration. While the control LNPs lacking targeting antibodies caused acute liver toxicity, trastuzumab-displayed LNPs showed no systemic toxicity. The tumor-specific delivery of p53 tumor suppressor mRNA led to the complete regression of cancer cells. Thus, apolipoprotein fusion enables a straightforward and scalable production of antibody-functionalized mRNA@LNPs, offering significant therapeutic potential in gene therapy.
Collapse
Affiliation(s)
- Wonbeom Park
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Jiwoong Choi
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
| | - Jaehyeon Hwang
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Suhyun Kim
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Yelee Kim
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
- Department
of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Man Kyu Shim
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
| | - Wooram Park
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Seokhyeon Yu
- Research
Center, MVRIX, Anyang 14058, Republic of Korea
| | - Sangwon Jung
- Research
Center, MVRIX, Anyang 14058, Republic of Korea
| | - Yoosoo Yang
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
- Division
of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Dae-Hyuk Kweon
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| |
Collapse
|
2
|
Haque MA, Shrestha A, Mikelis CM, Mattheolabakis G. Comprehensive analysis of lipid nanoparticle formulation and preparation for RNA delivery. Int J Pharm X 2024; 8:100283. [PMID: 39309631 PMCID: PMC11415597 DOI: 10.1016/j.ijpx.2024.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Nucleic acid-based therapeutics are a common approach that is increasingly popular for a wide spectrum of diseases. Lipid nanoparticles (LNPs) are promising delivery carriers that provide RNA stability, with strong transfection efficiency, favorable and tailorable pharmacokinetics, limited toxicity, and established translatability. In this review article, we describe the lipid-based delivery systems, focusing on lipid nanoparticles, the need of their use, provide a comprehensive analysis of each component, and highlight the advantages and disadvantages of the existing manufacturing processes. We further summarize the ongoing and completed clinical trials utilizing LNPs, indicating important aspects/questions worth of investigation, and analyze the future perspectives of this significant and promising therapeutic approach.
Collapse
Affiliation(s)
- Md. Anamul Haque
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Archana Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
3
|
Zhao S, Gao K, Han H, Stenzel M, Yin B, Song H, Lawanprasert A, Nielsen JE, Sharma R, Arogundade OH, Pimcharoen S, Chen YJ, Paul A, Tuma J, Collins MG, Wyle Y, Cranick MG, Burgstone BW, Perez BS, Barron AE, Smith AM, Lee HY, Wang A, Murthy N. Acid-degradable lipid nanoparticles enhance the delivery of mRNA. NATURE NANOTECHNOLOGY 2024; 19:1702-1711. [PMID: 39179796 DOI: 10.1038/s41565-024-01765-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 07/19/2024] [Indexed: 08/26/2024]
Abstract
Lipid nanoparticle (LNP)-mRNA complexes are transforming medicine. However, the medical applications of LNPs are limited by their low endosomal disruption rates, high toxicity and long tissue persistence times. LNPs that rapidly hydrolyse in endosomes (RD-LNPs) could solve the problems limiting LNP-based therapeutics and dramatically expand their applications but have been challenging to synthesize. Here we present an acid-degradable linker termed 'azido-acetal' that hydrolyses in endosomes within minutes and enables the production of RD-LNPs. Acid-degradable lipids composed of polyethylene glycol lipids, anionic lipids and cationic lipids were synthesized with the azido-acetal linker and used to generate RD-LNPs, which significantly improved the performance of LNP-mRNA complexes in vitro and in vivo. Collectively, RD-LNPs delivered mRNA more efficiently to the liver, lung, spleen and brains of mice and to haematopoietic stem and progenitor cells in vitro than conventional LNPs. These experiments demonstrate that engineering LNP hydrolysis rates in vivo has great potential for expanding the medical applications of LNPs.
Collapse
Affiliation(s)
- Sheng Zhao
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Kewa Gao
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Hesong Han
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA.
| | - Michael Stenzel
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Boyan Yin
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Hengyue Song
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Atip Lawanprasert
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Josefine Eilsø Nielsen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, CA, USA
| | - Rohit Sharma
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Opeyemi H Arogundade
- Department of Bioengineering and Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Sopida Pimcharoen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Yu-Ju Chen
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Abhik Paul
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jan Tuma
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, Plzen, Czech Republic
| | - Michael G Collins
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yofiel Wyle
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Matileen Grace Cranick
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Benjamin W Burgstone
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Barbara S Perez
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Annelise E Barron
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Andrew M Smith
- Department of Bioengineering and Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Hye Young Lee
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Aijun Wang
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA.
| | - Niren Murthy
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
4
|
Alameh MG, Tavakoli Naeini A, Dwivedi G, Lesage F, Buschmann MD, Lavertu M. Chitosan siRNA Nanoparticles Produce Significant Non-Toxic Functional Gene Silencing in Kidney Cortices. Polymers (Basel) 2024; 16:2547. [PMID: 39274180 PMCID: PMC11398103 DOI: 10.3390/polym16172547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024] Open
Abstract
Chitosan shows effective nucleic acid delivery. To understand the influence of chitosan's molecular weight, dose, payload, and hyaluronic acid coating on in vivo toxicity, immune stimulation, biodistribution and efficacy, precisely characterized chitosans were formulated with unmodified or chemically modified siRNA to control for innate immune stimulation. The hemocompatibility, cytokine induction, hematological and serological responses were assessed. Body weight, clinical signs, in vivo biodistribution and functional target knockdown were monitored. Hemolysis was found to be dose- and MW-dependent with the HA coating abrogating hemolysis. Compared to cationic lipid nanoparticles, uncoated and HA-coated chitosan nanoparticles did not induce immune stimulation or hematologic toxicity. Liver and kidney biomarkers remained unchanged with chitosan formulations, while high doses of cationic lipid nanoparticles led to increased transaminase levels and a decrease in body weight. Uncoated and HA-coated nanoparticles accumulated in kidneys with functional knockdown for uncoated chitosan formulations reaching 60%, suggesting potential applications in the treatment of kidney diseases.
Collapse
Affiliation(s)
- Mohamad-Gabriel Alameh
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Ashkan Tavakoli Naeini
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
| | - Garima Dwivedi
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
| | - Frederic Lesage
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Michael D Buschmann
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Marc Lavertu
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
5
|
Hsia T, Chen Y. RNA-encapsulating lipid nanoparticles in cancer immunotherapy: From pre-clinical studies to clinical trials. Eur J Pharm Biopharm 2024; 197:114234. [PMID: 38401743 DOI: 10.1016/j.ejpb.2024.114234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 02/26/2024]
Abstract
Nanoparticle-based delivery systems such as RNA-encapsulating lipid nanoparticles (RNA LNPs) have dramatically advanced in function and capacity over the last few decades. RNA LNPs boast of a diverse array of external and core configurations that enhance targeted delivery and prolong circulatory retention, advancing therapeutic outcomes. Particularly within the realm of cancer immunotherapies, RNA LNPs are increasingly gaining prominence. Pre-clinical in vitro and in vivo studies have laid a robust foundation for new and ongoing clinical trials that are actively enrolling patients for RNA LNP cancer immunotherapy. This review explores RNA LNPs, starting from their core composition to their external membrane formulation, set against a backdrop of recent clinical breakthroughs. We further elucidate the LNP delivery avenues, broach the prevailing challenges, and contemplate the future perspectives of RNA LNP-mediated immunotherapy.
Collapse
Affiliation(s)
- Tiffaney Hsia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
6
|
Kola NS, Patel D, Thakur A. RNA-Based Vaccines and Therapeutics Against Intracellular Pathogens. Methods Mol Biol 2024; 2813:321-370. [PMID: 38888787 DOI: 10.1007/978-1-0716-3890-3_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
RNA-based vaccines have sparked a paradigm shift in the treatment and prevention of diseases by nucleic acid medicines. There has been a notable surge in the development of nucleic acid therapeutics and vaccines following the global approval of the two messenger RNA-based COVID-19 vaccines. This growth is fueled by the exploration of numerous RNA products in preclinical stages, offering several advantages over conventional methods, i.e., safety, efficacy, scalability, and cost-effectiveness. In this chapter, we provide an overview of various types of RNA and their mechanisms of action for stimulating immune responses and inducing therapeutic effects. Furthermore, this chapter delves into the varying delivery systems, particularly emphasizing the use of nanoparticles to deliver RNA. The choice of delivery system is an intricate process involved in developing nucleic acid medicines that significantly enhances their stability, biocompatibility, and site-specificity. Additionally, this chapter sheds light on the current landscape of clinical trials of RNA therapeutics and vaccines against intracellular pathogens.
Collapse
Affiliation(s)
- Naga Suresh Kola
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Dhruv Patel
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Aneesh Thakur
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
7
|
Wang YS, Kumari M, Chen GH, Hong MH, Yuan JPY, Tsai JL, Wu HC. mRNA-based vaccines and therapeutics: an in-depth survey of current and upcoming clinical applications. J Biomed Sci 2023; 30:84. [PMID: 37805495 PMCID: PMC10559634 DOI: 10.1186/s12929-023-00977-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023] Open
Abstract
mRNA-based drugs have tremendous potential as clinical treatments, however, a major challenge in realizing this drug class will promise to develop methods for safely delivering the bioactive agents with high efficiency and without activating the immune system. With regard to mRNA vaccines, researchers have modified the mRNA structure to enhance its stability and promote systemic tolerance of antigenic presentation in non-inflammatory contexts. Still, delivery of naked modified mRNAs is inefficient and results in low levels of antigen protein production. As such, lipid nanoparticles have been utilized to improve delivery and protect the mRNA cargo from extracellular degradation. This advance was a major milestone in the development of mRNA vaccines and dispelled skepticism about the potential of this technology to yield clinically approved medicines. Following the resounding success of mRNA vaccines for COVID-19, many other mRNA-based drugs have been proposed for the treatment of a variety of diseases. This review begins with a discussion of mRNA modifications and delivery vehicles, as well as the factors that influence administration routes. Then, we summarize the potential applications of mRNA-based drugs and discuss further key points pertaining to preclinical and clinical development of mRNA drugs targeting a wide range of diseases. Finally, we discuss the latest market trends and future applications of mRNA-based drugs.
Collapse
Affiliation(s)
- Yu-Shiuan Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Monika Kumari
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Guan-Hong Chen
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Ming-Hsiang Hong
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Joyce Pei-Yi Yuan
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Jui-Ling Tsai
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan.
| |
Collapse
|
8
|
Bernal-Martínez AM, Angulo-Pachón CA, Galindo F, Miravet JF. Reduction-Responsive Cationic Vesicles from Bolaamphiphiles with Ionizable Amino Acid or Dipeptide Polar Heads. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:13841-13849. [PMID: 37729523 PMCID: PMC10552552 DOI: 10.1021/acs.langmuir.3c01294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/31/2023] [Indexed: 09/22/2023]
Abstract
This paper presents a study of the aggregation of cationic bolaamphiphilic molecules into vesicles. These molecules are based on a cystamine core with protonated terminal dipeptide groups. The study found that vesicles can be formed at pH 4 for all of the dipeptide-terminated bolaamphiphiles containing different combinations of l-valine, l-phenylalanine, and l-tryptophan. The concentration for aggregation onset was determined by using pyrene as a fluorescent probe or light dispersion for compounds with tryptophan. Dynamic light scattering (DLS) studies and transmission electron microscopy (TEM) reveal that the vesicles have diameters ranging from 140 to 500 nm and show the capability of loading hydrophobic cargos, such as Nile red, and their liberation in reductive environments. Furthermore, the bolaamphiphiles are only fully protonated and prone to vesicle formation at acidic pH, making them a promising alternative for gastrointestinal delivery.
Collapse
Affiliation(s)
- Ana M. Bernal-Martínez
- Department of Inorganic and
Organic Chemistry, Universitat Jaume I, 12071 Castelló
de la Plana, Spain
| | - César A. Angulo-Pachón
- Department of Inorganic and
Organic Chemistry, Universitat Jaume I, 12071 Castelló
de la Plana, Spain
| | - Francisco Galindo
- Department of Inorganic and
Organic Chemistry, Universitat Jaume I, 12071 Castelló
de la Plana, Spain
| | - Juan F. Miravet
- Department of Inorganic and
Organic Chemistry, Universitat Jaume I, 12071 Castelló
de la Plana, Spain
| |
Collapse
|
9
|
Jeong M, Lee Y, Park J, Jung H, Lee H. Lipid nanoparticles (LNPs) for in vivo RNA delivery and their breakthrough technology for future applications. Adv Drug Deliv Rev 2023; 200:114990. [PMID: 37423563 DOI: 10.1016/j.addr.2023.114990] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
RNA therapeutics show a significant breakthrough for the treatment of otherwise incurable diseases and genetic disorders by regulating disease-related gene expression. The successful development of COVID-19 mRNA vaccines further emphasizes the potential of RNA therapeutics in the prevention of infectious diseases as well as in the treatment of chronic diseases. However, the efficient delivery of RNA into cells remains a challenge, and nanoparticle delivery systems such as lipid nanoparticles (LNPs) are necessary to fully realize the potential of RNA therapeutics. While LNPs provide a highly efficient platform for the in vivo delivery of RNA by overcoming various biological barriers, several challenges remain to be resolved for further development and regulatory approval. These include a lack of targeted delivery to extrahepatic organs and a gradual loss of therapeutic potency with repeated doses. In this review, we highlight the fundamental aspects of LNPs and their uses in the development of novel RNA therapeutics. Recent advances in LNP-based therapeutics and preclinical/clinical studies are overviewed. Lastly, we discuss the current limitations of LNPs and introduce breakthrough technologies that might overcome these challenges in future applications.
Collapse
Affiliation(s)
- Michaela Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Yeji Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Jeongeun Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Hyein Jung
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea.
| |
Collapse
|
10
|
Liu C, Shi Q, Huang X, Koo S, Kong N, Tao W. mRNA-based cancer therapeutics. Nat Rev Cancer 2023:10.1038/s41568-023-00586-2. [PMID: 37311817 DOI: 10.1038/s41568-023-00586-2] [Citation(s) in RCA: 155] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 06/15/2023]
Abstract
Due to the fact that mRNA technology allows the production of diverse vaccines and treatments in a shorter time frame and with reduced expense compared to conventional approaches, there has been a surge in the use of mRNA-based therapeutics in recent years. With the aim of encoding tumour antigens for cancer vaccines, cytokines for immunotherapy, tumour suppressors to inhibit tumour development, chimeric antigen receptors for engineered T cell therapy or genome-editing proteins for gene therapy, many of these therapeutics have shown promising efficacy in preclinical studies, and some have even entered clinical trials. Given the evidence supporting the effectiveness and safety of clinically approved mRNA vaccines, coupled with growing interest in mRNA-based therapeutics, mRNA technology is poised to become one of the major pillars in cancer drug development. In this Review, we present in vitro transcribed mRNA-based therapeutics for cancer treatment, including the characteristics of the various types of synthetic mRNA, the packaging systems for efficient mRNA delivery, preclinical and clinical studies, current challenges and future prospects in the field. We anticipate the translation of promising mRNA-based treatments into clinical applications, to ultimately benefit patients.
Collapse
Affiliation(s)
- Chuang Liu
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Qiangqiang Shi
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Xiangang Huang
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Protein Transduction Domain-Mediated Delivery of Recombinant Proteins and In Vitro Transcribed mRNAs for Protein Replacement Therapy of Human Severe Genetic Mitochondrial Disorders: The Case of Sco2 Deficiency. Pharmaceutics 2023; 15:pharmaceutics15010286. [PMID: 36678915 PMCID: PMC9861957 DOI: 10.3390/pharmaceutics15010286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/31/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
Mitochondrial disorders represent a heterogeneous group of genetic disorders with variations in severity and clinical outcomes, mostly characterized by respiratory chain dysfunction and abnormal mitochondrial function. More specifically, mutations in the human SCO2 gene, encoding the mitochondrial inner membrane Sco2 cytochrome c oxidase (COX) assembly protein, have been implicated in the mitochondrial disorder fatal infantile cardioencephalomyopathy with COX deficiency. Since an effective treatment is still missing, a protein replacement therapy (PRT) was explored using protein transduction domain (PTD) technology. Therefore, the human recombinant full-length mitochondrial protein Sco2, fused to TAT peptide (a common PTD), was produced (fusion Sco2 protein) and successfully transduced into fibroblasts derived from a SCO2/COX-deficient patient. This PRT contributed to effective COX assembly and partial recovery of COX activity. In mice, radiolabeled fusion Sco2 protein was biodistributed in the peripheral tissues of mice and successfully delivered into their mitochondria. Complementary to that, an mRNA-based therapeutic approach has been more recently considered as an innovative treatment option. In particular, a patented, novel PTD-mediated IVT-mRNA delivery platform was developed and applied in recent research efforts. PTD-IVT-mRNA of full-length SCO2 was successfully transduced into the fibroblasts derived from a SCO2/COX-deficient patient, translated in host ribosomes into a nascent chain of human Sco2, imported into mitochondria, and processed to the mature protein. Consequently, the recovery of reduced COX activity was achieved, thus suggesting the potential of this mRNA-based technology for clinical translation as a PRT for metabolic/genetic disorders. In this review, such research efforts will be comprehensibly presented and discussed to elaborate their potential in clinical application and therapeutic usefulness.
Collapse
|
12
|
Gómez-Aguado I, Rodríguez-Castejón J, Beraza-Millor M, Rodríguez-Gascón A, Del Pozo-Rodríguez A, Solinís MÁ. mRNA delivery technologies: Toward clinical translation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 372:207-293. [PMID: 36064265 DOI: 10.1016/bs.ircmb.2022.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Messenger RNA (mRNA)-therapies have recently taken a huge step toward clinic thanks to the first mRNA-based medicinal products marketed. mRNA features for clinical purposes are improved by chemical modifications, but the inclusion in a delivery system is a regular requirement. mRNA nanomedicines must be designed for the specific therapeutic purpose, protecting the nucleic acid and facilitating the overcoming of biological barriers. Polymers, polypeptides, and cationic lipids are the main used materials to design mRNA delivery systems. Among them, lipid nanoparticles (LNPs) are the most advanced ones, and currently they are at the forefront of preclinical and clinical evaluation in several fields, including immunotherapy (against infectious diseases and cancer), protein replacement, gene editing and regenerative medicine. This chapter includes an overview on mRNA delivery technologies, with special interest in LNPs, and the most recent advances in their clinical application. Liposomes are the mRNA delivery technology with the highest clinical translation among LNPs, whereas the first clinical trial of a therapeutic mRNA formulated in exosomes has been recently approved for protein replacement therapy. The first mRNA products approved by the regulatory agencies worldwide are LNP-based mRNA vaccines against viral infections, specifically against the 2019 coronavirus disease (COVID-19). The clinical translation of mRNA-therapies for cancer is mainly focused on three strategies: anti-cancer vaccination by means of delivering cancer antigens or acting as an adjuvant, mRNA-engineered chimeric antigen receptors (CARs) and T-cell receptors (TCRs), and expression of antibodies and immunomodulators. Cancer immunotherapy and, more recently, COVID-19 vaccines spearhead the advance of mRNA clinical use.
Collapse
Affiliation(s)
- Itziar Gómez-Aguado
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Julen Rodríguez-Castejón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Marina Beraza-Millor
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Alicia Rodríguez-Gascón
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - Ana Del Pozo-Rodríguez
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain
| | - María Ángeles Solinís
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de investigación Lascaray ikergunea, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, Vitoria-Gasteiz, Spain.
| |
Collapse
|
13
|
Ghidini M, Silva SG, Evangelista J, do Vale MLC, Farooqi AA, Pinheiro M. Nanomedicine for the Delivery of RNA in Cancer. Cancers (Basel) 2022; 14:2677. [PMID: 35681657 PMCID: PMC9179531 DOI: 10.3390/cancers14112677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023] Open
Abstract
The complexity, and the diversity of the different types of cancers allied to the tendency to form metastasis make treatment efficiency so tricky and often impossible due to the advanced stage of the disease in the diagnosis. In recent years, due to tremendous scientific breakthroughs, we have witnessed exponential growth in the elucidation of mechanisms that underlie carcinogenesis and metastasis. The development of more selective therapies made it possible to improve cancer treatment. Although interdisciplinary research leads to encouraging results, scientists still have a long exploration journey. RNA technology represents a promise as a therapeutic intervention for targeted gene silencing in cancer, and there are already some RNA-based formulations in clinical trials. However, the use of RNA as a therapeutic tool presents severe limitations, mainly related to its low stability and poor cellular uptake. Thus, the use of nanomedicine employing nanoparticles to encapsulate RNA may represent a suitable platform to address the major challenges hampering its therapeutic application. In this review, we have revisited the potential of RNA and RNA-associated therapies to fight cancer, also providing, as support, a general overview of nanoplatforms for RNA delivery.
Collapse
Affiliation(s)
- Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Sandra G. Silva
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal; (S.G.S.); (M.L.C.d.V.)
| | - Jessica Evangelista
- Thoracic Surgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
| | - Maria Luísa C. do Vale
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal; (S.G.S.); (M.L.C.d.V.)
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan;
| | - Marina Pinheiro
- REQUIMTE, University of Porto, 4169-007 Porto, Portugal
- ICVS, Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
14
|
Wisitrasameewong W, Champaiboon C, Surisaeng T, Sa-Ard-Iam N, Freire M, Pardi N, Pichyangkul S, Mahanonda R. The Impact of mRNA Technology in Regenerative Therapy: Lessons for Oral Tissue Regeneration. J Dent Res 2022; 101:1015-1024. [PMID: 35319289 DOI: 10.1177/00220345221084205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Oral tissue regeneration following chronic diseases and injuries is limited by the natural endogenous wound-healing process. Current regenerative approaches implement exogenous systems, including stem cells, scaffolds, growth factors, and plasmid DNA/viral vectors, that induce variable clinical outcomes. An innovative approach that is safe, effective, and inexpensive is needed. The lipid nanoparticle-encapsulated nucleoside-modified messenger RNA (mRNA) platform has proven to be a successful vaccine modality against coronavirus disease 2019, demonstrating safety and high efficacy in humans. The same fundamental technology platform could be applied to facilitate the development of mRNA-based regenerative therapy. While the platform has not yet been studied in the field of oral tissue regeneration, mRNA therapeutics encoding growth factors have been evaluated and demonstrated promising findings in various models of soft and hard tissue regeneration such as myocardial infarction, diabetic wound healing, and calvarial and femoral bone defects. Because restoration of both soft and hard tissues is crucial to oral tissue physiology, this new therapeutic modality may help to overcome challenges associated with the reconstruction of the unique and complex architecture of oral tissues. This review discusses mRNA therapeutics with an emphasis on findings and lessons in different regenerative animal models, and it speculates how we can apply mRNA-based platforms for oral tissue regeneration.
Collapse
Affiliation(s)
- W Wisitrasameewong
- Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Center of Excellence in Periodontal Disease and Dental Implant, Chulalongkorn University, Bangkok, Thailand.,Immunology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - C Champaiboon
- Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Center of Excellence in Periodontal Disease and Dental Implant, Chulalongkorn University, Bangkok, Thailand.,Immunology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - T Surisaeng
- Department of Conservative Dentistry, Faculty of Dentistry, Prince of Songkhla University, Songkhla, Thailand
| | - N Sa-Ard-Iam
- Center of Excellence in Periodontal Disease and Dental Implant, Chulalongkorn University, Bangkok, Thailand.,Immunology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - M Freire
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, USA
| | - N Pardi
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - S Pichyangkul
- Department of Bacterial and Parasitic Diseases, AFRIMS, Bangkok, Thailand
| | - R Mahanonda
- Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Center of Excellence in Periodontal Disease and Dental Implant, Chulalongkorn University, Bangkok, Thailand.,Immunology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
15
|
Xu J, Wan K, Wang H, Shi X, Wang J, Zhong Y, Gao C, Zhang Y, Nie G. Polyethylenimine-Poly(lactic- co-glycolic acid) 2 Nanoparticles Show an Innate Targeting Ability to the Submandibular Salivary Gland via the Muscarinic 3 Receptor. ACS CENTRAL SCIENCE 2021; 7:1938-1948. [PMID: 34841064 PMCID: PMC8614106 DOI: 10.1021/acscentsci.1c01083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Indexed: 06/13/2023]
Abstract
Polymeric nanoparticles have been extensively explored for biomedical applications, especially as framework materials for the construction of functional nanostructures. However, less attention has been paid to the inherent biological activities of those polymers. In this work, one of the commonly used polymers in gene and protein delivery, polyethylenimine-poly(lactic-co-glycolic acid)2 (PEI-PLGA), was discovered by accident to be able to mediate the nanoparticles to target the submandibular salivary glands of mice after intravenous injection. PEI-PLGA nanoparticles with an unmodified PEI surface selectively accumulated in submandibular salivary glands with ex vivo and in vitro study, suggesting that a ligand-receptor interaction between PEI and muscarinic acetylcholine receptor subtype 3 (M3 receptor) contributed to this affinity. Docking computation for the molecular binding mode between PEI segments and M3 receptor indicated the way they interacted was similar to that of the FDA-approved specific M3 receptor antagonist, tiotropium. The key amino acids mediated this specific interaction between PEI-PLGA nanoparticles and M3 receptor were identified via a simulated alanine mutation study. This work demonstrates the unique characteristic of PEI-PLGA nanoparticles, which may be useful for the development of muscarinic receptor targeted nanomedicines and should be taken into consideration when PEI-based nanoparticles are applied in gene delivery.
Collapse
Affiliation(s)
- Junchao Xu
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety,
Laboratory of Theoretical and Computational Nanoscience, CAS Center
for Excellence in Nanoscience, National
Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Kaiwei Wan
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety,
Laboratory of Theoretical and Computational Nanoscience, CAS Center
for Excellence in Nanoscience, National
Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hui Wang
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety,
Laboratory of Theoretical and Computational Nanoscience, CAS Center
for Excellence in Nanoscience, National
Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xinghua Shi
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety,
Laboratory of Theoretical and Computational Nanoscience, CAS Center
for Excellence in Nanoscience, National
Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jing Wang
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety,
Laboratory of Theoretical and Computational Nanoscience, CAS Center
for Excellence in Nanoscience, National
Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yi Zhong
- State
Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical
Sciences, Peking University, Beijing 100191, China
| | - Chao Gao
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety,
Laboratory of Theoretical and Computational Nanoscience, CAS Center
for Excellence in Nanoscience, National
Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yinlong Zhang
- School
of Nanoscience and Technology, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangjun Nie
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety,
Laboratory of Theoretical and Computational Nanoscience, CAS Center
for Excellence in Nanoscience, National
Center for Nanoscience and Technology, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
16
|
Ibba ML, Ciccone G, Esposito CL, Catuogno S, Giangrande PH. Advances in mRNA non-viral delivery approaches. Adv Drug Deliv Rev 2021; 177:113930. [PMID: 34403751 DOI: 10.1016/j.addr.2021.113930] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/28/2021] [Accepted: 08/11/2021] [Indexed: 12/11/2022]
Abstract
Messenger RNAs (mRNAs) present a great potential as therapeutics for the treatment and prevention of a wide range of human pathologies, allowing for protein replacement, vaccination, cancer immunotherapy, and genomic engineering. Despite advances in the design of mRNA-based therapeutics, a key aspect for their widespread translation to clinic is the development of safe and effective delivery strategies. To this end, non-viral delivery systems including peptide-based complexes, lipidic or polymeric nanoparticles, and hybrid formulations are attracting growing interest. Despite displaying somewhat reduced efficacy compared to viral-based systems, non-viral carriers offer important advantages in terms of biosafety and versatility. In this review, we provide an overview of current mRNA therapeutic applications and discuss key biological barriers to delivery and recent advances in the development of non-viral systems. Challenges and future applications of this novel therapeutic modality are also discussed.
Collapse
Affiliation(s)
- Maria L Ibba
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, 80131 Naples, Italy
| | - Giuseppe Ciccone
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), 80145 Naples, Italy
| | - Carla L Esposito
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), 80145 Naples, Italy.
| | - Silvia Catuogno
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), 80145 Naples, Italy.
| | - Paloma H Giangrande
- University of Iowa, Department of Internal Medicine, Iowa City, IA, USA; Wave Life Sciences, Cambridge, MA, USA.
| |
Collapse
|
17
|
Bost JP, Barriga H, Holme MN, Gallud A, Maugeri M, Gupta D, Lehto T, Valadi H, Esbjörner EK, Stevens MM, El-Andaloussi S. Delivery of Oligonucleotide Therapeutics: Chemical Modifications, Lipid Nanoparticles, and Extracellular Vesicles. ACS NANO 2021; 15:13993-14021. [PMID: 34505766 PMCID: PMC8482762 DOI: 10.1021/acsnano.1c05099] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Indexed: 05/04/2023]
Abstract
Oligonucleotides (ONs) comprise a rapidly growing class of therapeutics. In recent years, the list of FDA-approved ON therapies has rapidly expanded. ONs are small (15-30 bp) nucleotide-based therapeutics which are capable of targeting DNA and RNA as well as other biomolecules. ONs can be subdivided into several classes based on their chemical modifications and on the mechanisms of their target interactions. Historically, the largest hindrance to the widespread usage of ON therapeutics has been their inability to effectively internalize into cells and escape from endosomes to reach their molecular targets in the cytosol or nucleus. While cell uptake has been improved, "endosomal escape" remains a significant problem. There are a range of approaches to overcome this, and in this review, we focus on three: altering the chemical structure of the ONs, formulating synthetic, lipid-based nanoparticles to encapsulate the ONs, or biologically loading the ONs into extracellular vesicles. This review provides a background to the design and mode of action of existing FDA-approved ONs. It presents the most common ON classifications and chemical modifications from a fundamental scientific perspective and provides a roadmap of the cellular uptake pathways by which ONs are trafficked. Finally, this review delves into each of the above-mentioned approaches to ON delivery, highlighting the scientific principles behind each and covering recent advances.
Collapse
Affiliation(s)
- Jeremy P. Bost
- Department
of Laboratory Medicine, Karolinska Institutet, Huddinge 14152, Sweden
| | - Hanna Barriga
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 17177, Sweden
| | - Margaret N. Holme
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 17177, Sweden
| | - Audrey Gallud
- Department
of Biology and Biological Engineering, Chalmers
University of Technology, Gothenburg 41296, Sweden
- Advanced
Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg 43150, Sweden
| | - Marco Maugeri
- Department
of Rheumatology and Inflammation Research, Institute of Medicine,
Sahlgrenska Academy, University of Gothenburg, Gothenburg 41390, Sweden
| | - Dhanu Gupta
- Department
of Laboratory Medicine, Karolinska Institutet, Huddinge 14152, Sweden
| | - Taavi Lehto
- Department
of Laboratory Medicine, Karolinska Institutet, Huddinge 14152, Sweden
- Institute
of Technology, University of Tartu, Nooruse 1, Tartu 50411, Estonia
| | - Hadi Valadi
- Department
of Rheumatology and Inflammation Research, Institute of Medicine,
Sahlgrenska Academy, University of Gothenburg, Gothenburg 41390, Sweden
| | - Elin K. Esbjörner
- Department
of Biology and Biological Engineering, Chalmers
University of Technology, Gothenburg 41296, Sweden
| | - Molly M. Stevens
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 17177, Sweden
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London SW7 2BU, United Kingdom
| | - Samir El-Andaloussi
- Department
of Laboratory Medicine, Karolinska Institutet, Huddinge 14152, Sweden
| |
Collapse
|
18
|
mRNA delivery via non-viral carriers for biomedical applications. Int J Pharm 2021; 607:121020. [PMID: 34416327 DOI: 10.1016/j.ijpharm.2021.121020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/03/2021] [Accepted: 08/14/2021] [Indexed: 12/11/2022]
Abstract
As an emerging new class of nucleic acid drugs, messenger RNA (mRNA) has huge potential in immunotherapy, regenerative medicine, vaccine, and gene editing. Comparing with siRNA and pDNA, mRNA is more vulnerable to nucleases in vivo. However, the lack of effective and safe delivery methods impedes the broad application of mRNA-based therapeutics. Up to now, the delivery of mRNA remains largely unexplored, and therefore, is a hot topic in the field of gene therapy. In this review, we will summarize the ongoing challenges in mRNA-based therapeutics and unmet requirements for delivery vehicles in terms of the unique structure of mRNA. We then highlight the advancement in mRNA delivery in both fundamental research and clinical applications. Finally, a prospective will be proposed upon reviewing the current progress in mRNA delivery.
Collapse
|
19
|
Granados-Riveron JT, Aquino-Jarquin G. Engineering of the current nucleoside-modified mRNA-LNP vaccines against SARS-CoV-2. Biomed Pharmacother 2021; 142:111953. [PMID: 34343897 PMCID: PMC8299225 DOI: 10.1016/j.biopha.2021.111953] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Currently, there are over 230 different COVID-19 vaccines under development around the world. At least three decades of scientific development in RNA biology, immunology, structural biology, genetic engineering, chemical modification, and nanoparticle technologies allowed the accelerated development of fully synthetic messenger RNA (mRNA)-based vaccines within less than a year since the first report of a SARS-CoV-2 infection. mRNA-based vaccines have been shown to elicit broadly protective immune responses, with the added advantage of being amenable to rapid and flexible manufacturing processes. This review recapitulates current advances in engineering the first two SARS-CoV-2-spike-encoding nucleoside-modified mRNA vaccines, highlighting the strategies followed to potentiate their effectiveness and safety, thus facilitating an agile response to the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Javier T Granados-Riveron
- Laboratorio de Investigación en Patogénesis Molecular, Hospital Infantil de México, Federico Gómez, Ciudad de México, Mexico
| | - Guillermo Aquino-Jarquin
- Laboratorio de Investigación en Genómica, Genética y Bioinformática, Hospital Infantil de México, Federico Gómez, Ciudad de México, Mexico.
| |
Collapse
|
20
|
Fu X, Chen T, Song Y, Feng C, Chen H, Zhang Q, Chen G, Zhu X. mRNA Delivery by a pH-Responsive DNA Nano-Hydrogel. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101224. [PMID: 34145748 DOI: 10.1002/smll.202101224] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 06/12/2023]
Abstract
The delivery of mRNA to manipulate protein expression has attracted widespread attention, since that mRNA overcomes the problem of infection and mutation risks in transgenes and can work as drugs for the treatment of diseases. Although there are currently some vehicles that deliver mRNA into cells, they have not yet reached a good balance in terms of expression efficiency and biocompatibility. Here, a DNA nano-hydrogel system for mRNA delivery is developed. The nano-hydrogel is all composed of DNA except the target mRNA, so it has superior biocompatibility compared with those chemical vehicles. In parallel, the nano-hydrogel can be compacted into a nanosphere under the crosslinking by well-designed "X"-shaped DNA scaffolds and DNA linkers, facilitating the delivery into cells through endocytosis. In addition, smart intracellular release of the mRNA is achieved by incorporating a pH-responsive i-motif structure into the nano-hydrogel. Thus, taking the efficient delivery and release together, mRNA can be translated into the corresponding protein with a high efficiency, which is comparable to that of the commercial liposome but with a much better biocompatibility. Due to the excellent biocompatibility and efficiency, this nano-hydrogel system is expected to become a competitive alternative for delivering functional mRNA in vivo.
Collapse
Affiliation(s)
- Xin Fu
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Tianshu Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yuchen Song
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Chang Feng
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Huinan Chen
- College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Qianqian Zhang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Guifang Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiaoli Zhu
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
21
|
Chen F, Wang Y, Gao J, Saeed M, Li T, Wang W, Yu H. Nanobiomaterial-based vaccination immunotherapy of cancer. Biomaterials 2021; 270:120709. [PMID: 33581608 DOI: 10.1016/j.biomaterials.2021.120709] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/27/2021] [Accepted: 01/31/2021] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapies including cancer vaccines, immune checkpoint blockade or chimeric antigen receptor T cells have been exploited as the attractive treatment modalities in recent years. Among these approaches, cancer vaccines that designed to deliver tumor antigens and adjuvants to activate the antigen presenting cells (APCs) and induce antitumor immune responses, have shown significant efficacy in inhibiting tumor growth, preventing tumor relapse and metastasis. Despite the potential of cancer vaccination strategies, the therapeutic outcomes in preclinical trials are failed to promote their clinical translation, which is in part due to their inefficient vaccination cascade of five critical steps: antigen identification, antigen encapsulation, antigen delivery, antigen release and antigen presentation to T cells. In recent years, it has been demonstrated that various nanobiomaterials hold great potential to enhance cancer vaccination cascade and improve their antitumor performance and reduce the off-target effect. We summarize the cutting-edge advances of nanobiomaterials-based vaccination immunotherapy of cancer in this review. The various cancer nanovaccines including antigen peptide/adjuvant-based nanovaccines, nucleic acid-based nanovaccines as well as biomimetic nanobiomaterials-based nanovaccines are discussed in detail. We also provide some challenges and perspectives associated with the clinical translation of cancer nanovaccines.
Collapse
Affiliation(s)
- Fangmin Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingjie Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Nano Science and Technology Institute, University of Science and Technology of China, Suzhou, 215123, China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Madiha Saeed
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tianliang Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiqi Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
22
|
Karpenko LI, Rudometov AP, Sharabrin SV, Shcherbakov DN, Borgoyakova MB, Bazhan SI, Volosnikova EA, Rudometova NB, Orlova LA, Pyshnaya IA, Zaitsev BN, Volkova NV, Azaev MS, Zaykovskaya AV, Pyankov OV, Ilyichev AA. Delivery of mRNA Vaccine against SARS-CoV-2 Using a Polyglucin:Spermidine Conjugate. Vaccines (Basel) 2021; 9:76. [PMID: 33494530 PMCID: PMC7910849 DOI: 10.3390/vaccines9020076] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
One of the key stages in the development of mRNA vaccines is their delivery. Along with liposome, other materials are being developed for mRNA delivery that can ensure both the safety and effectiveness of the vaccine, and also facilitate its storage and transportation. In this study, we investigated the polyglucin:spermidine conjugate as a carrier of an mRNA-RBD vaccine encoding the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. The conditions for the self-assembling of mRNA-PGS complexes were optimized, including the selection of the mRNA:PGS charge ratios. Using dynamic and electrophoretic light scattering it was shown that the most monodisperse suspension of nanoparticles was formed at the mRNA:PGS charge ratio equal to 1:5. The average hydrodynamic particles diameter was determined, and it was confirmed by electron microscopy. The evaluation of the zeta potential of the investigated complexes showed that the particles surface charge was close to the zero point. This may indicate that the positively charged PGS conjugate has completely packed the negatively charged mRNA molecules. It has been shown that the packaging of mRNA-RBD into the PGS envelope leads to increased production of specific antibodies with virus-neutralizing activity in immunized BALB/c mice. Our results showed that the proposed polycationic polyglucin:spermidine conjugate can be considered a promising and safe means to the delivery of mRNA vaccines, in particular mRNA vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Larisa I. Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Andrey P. Rudometov
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Sergei V. Sharabrin
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Dmitry N. Shcherbakov
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Mariya B. Borgoyakova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Sergei I. Bazhan
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Ekaterina A. Volosnikova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Nadezhda B. Rudometova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Lyubov A. Orlova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Inna A. Pyshnaya
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Boris N. Zaitsev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Natalya V. Volkova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Mamedyar Sh. Azaev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Anna V. Zaykovskaya
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Oleg V. Pyankov
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| | - Alexander A. Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk, Russia; (A.P.R.); (S.V.S.); (D.N.S.); (M.B.B.); (S.I.B.); (E.A.V.); (N.B.R.); (L.A.O.); (B.N.Z.); (N.V.V.); (M.S.A.); (A.V.Z.); (O.V.P.); (A.A.I.)
| |
Collapse
|
23
|
Figueroa-Espada CG, Hofbauer S, Mitchell MJ, Riley RS. Exploiting the placenta for nanoparticle-mediated drug delivery during pregnancy. Adv Drug Deliv Rev 2020; 160:244-261. [PMID: 32956719 DOI: 10.1016/j.addr.2020.09.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/07/2020] [Accepted: 09/15/2020] [Indexed: 12/14/2022]
Abstract
A major challenge to treating diseases during pregnancy is that small molecule therapeutics are transported through the placenta and incur toxicities to the developing fetus. The placenta is responsible for providing nutrients, removing waste, and protecting the fetus from toxic substances. Thus, the placenta acts as a biological barrier between the mother and fetus that can be exploited for drug delivery. Nanoparticle technologies provide the opportunity for safe drug delivery during pregnancy by controlling how therapeutics interact with the placenta. In this Review, we present nanoparticle drug delivery technologies specifically designed to exploit the placenta as a biological barrier to treat maternal, placental, or fetal diseases exclusively, while minimizing off-target toxicities. Further, we discuss opportunities, challenges, and future directions for implementing drug delivery technologies during pregnancy.
Collapse
|
24
|
Affiliation(s)
- Chaoyang Meng
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Xiangya Hospital of Central South University Changsha Hunan 410000 China
| | - Zhe Chen
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Xiangya Hospital of Central South University Changsha Hunan 410000 China
| | - Gang Li
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
| | - Thomas Welte
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
| | - Haifa Shen
- Department of Nanomedicine Houston Methodist Research Institute Houston TX 77030 USA
- Cancer Center Houston Methodist Hospital Houston TX 77030 USA
- Department of Cell and Developmental Biology Weill Cornell Medical College New York NY 10065 USA
| |
Collapse
|
25
|
Lung P, Yang J, Li Q. Nanoparticle formulated vaccines: opportunities and challenges. NANOSCALE 2020; 12:5746-5763. [PMID: 32124894 DOI: 10.1039/c9nr08958f] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Vaccines harness the inherent properties of the immune system to prevent diseases or treat existing ones. Continuous efforts have been devoted to both gaining a mechanistic understanding of how the immune system operates and designing vaccines with high efficacies and effectiveness. Advancements in nanotechnology in recent years have generated unique opportunities to meet the daunting challenges associated with immunology and vaccine development. Firstly, nanoparticle formulated systems provide ideal model systems for studying the operation of the immune system, making it possible to systematically identify key factors and understand their roles in specific immune responses. Also, the versatile compositions/architectures of nanoparticle systems enable new strategies/novel platforms for developing vaccines with high efficacies and effectiveness. In this review, we discuss the advantages of nanoparticles and the challenges faced during vaccine development, through the framework of the immunological mechanisms of vaccination, with the aim of bridging the gap between immunology and materials science, which are both involved in vaccine design. The knowledge obtained provides general guidelines for future vaccine development.
Collapse
Affiliation(s)
- Pingsai Lung
- Department of Physics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | | | | |
Collapse
|
26
|
Wadhwa A, Aljabbari A, Lokras A, Foged C, Thakur A. Opportunities and Challenges in the Delivery of mRNA-based Vaccines. Pharmaceutics 2020; 12:E102. [PMID: 32013049 PMCID: PMC7076378 DOI: 10.3390/pharmaceutics12020102] [Citation(s) in RCA: 336] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/22/2020] [Accepted: 01/26/2020] [Indexed: 02/08/2023] Open
Abstract
In the past few years, there has been increasing focus on the use of messenger RNA (mRNA) as a new therapeutic modality. Current clinical efforts encompassing mRNA-based drugs are directed toward infectious disease vaccines, cancer immunotherapies, therapeutic protein replacement therapies, and treatment of genetic diseases. However, challenges that impede the successful translation of these molecules into drugs are that (i) mRNA is a very large molecule, (ii) it is intrinsically unstable and prone to degradation by nucleases, and (iii) it activates the immune system. Although some of these challenges have been partially solved by means of chemical modification of the mRNA, intracellular delivery of mRNA still represents a major hurdle. The clinical translation of mRNA-based therapeutics requires delivery technologies that can ensure stabilization of mRNA under physiological conditions. Here, we (i) review opportunities and challenges in the delivery of mRNA-based therapeutics with a focus on non-viral delivery systems, (ii) present the clinical status of mRNA vaccines, and (iii) highlight perspectives on the future of this promising new type of medicine.
Collapse
Affiliation(s)
| | | | | | | | - Aneesh Thakur
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| |
Collapse
|
27
|
Toy R, Pradhan P, Ramesh V, Di Paolo NC, Lash B, Liu J, Blanchard EL, Pinelli CJ, Santangelo PJ, Shayakhmetov DM, Roy K. Modification of primary amines to higher order amines reduces in vivo hematological and immunotoxicity of cationic nanocarriers through TLR4 and complement pathways. Biomaterials 2019; 225:119512. [PMID: 31585233 DOI: 10.1016/j.biomaterials.2019.119512] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/08/2019] [Accepted: 09/20/2019] [Indexed: 12/23/2022]
Abstract
For decades, cationic polymer nanoparticles have been investigated for nucleic acid delivery. Despite promising in vitro transfection results, most formulations have failed to translate into the clinic due to significant in vivo toxicity - especially when delivered intravenously. To address this significant problem, we investigated the detailed mechanisms that govern the complex in vivo systemic toxicity response to common polymeric nanoparticles. We determined that the toxicity response is material dependent. For branched polyethylenimine (bPEI) nanoparticles - toxicity is a function of multiple pathophysiological responses - triggering of innate immune sensors, induction of hepatic toxicity, and significant alteration of hematological properties. In contrast, for chitosan-based nanoparticles - systemic toxicity is primarily driven through innate immune activation. We further identified that modification of primary amines to secondary and tertiary amines using the small molecule imidazole-acetic-acid (IAA) ameliorates in vivo toxicity from both nanocarriers by different, material-specific mechanisms related to Toll-like receptor 4 activation (for bPEI) and complement activation driven neutrophil infiltration (for chitosan), respectively. Our results provide a detailed roadmap for evaluating in vivo toxicity of nanocarriers and identifies potential opportunities to reduce toxicity for eventual clinical translation.
Collapse
Affiliation(s)
- Randall Toy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia
| | - Pallab Pradhan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia
| | | | - Nelson C Di Paolo
- Department of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Blake Lash
- Wallace H. Coulter Department of Biomedical Engineering, Georgia
| | - Jiaying Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia
| | | | | | | | - Dmitry M Shayakhmetov
- Department of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia; Center for ImmunoEngineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
28
|
Therapeutic mRNA delivery to leukocytes. J Control Release 2019; 305:165-175. [DOI: 10.1016/j.jconrel.2019.05.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/15/2019] [Accepted: 05/19/2019] [Indexed: 12/14/2022]
|
29
|
Moss KH, Popova P, Hadrup SR, Astakhova K, Taskova M. Lipid Nanoparticles for Delivery of Therapeutic RNA Oligonucleotides. Mol Pharm 2019; 16:2265-2277. [PMID: 31063396 DOI: 10.1021/acs.molpharmaceut.8b01290] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gene therapy is an exciting field that has the potential to address emerging scientific and therapeutic tasks. RNA-based gene therapy has made remarkable progress in recent decades. Nevertheless, efficient targeted delivery of RNA therapeutics is still a prerequisite for entering the clinics. In this review, we introduce current delivery methods for RNA gene therapeutics based on lipid nanoparticles (LNPs). We focus on the clinical appeal of recent RNA NPs and discuss existing challenges of fabrication and screening LNP candidates for effective translation into drugs of human metabolic diseases and cancer.
Collapse
Affiliation(s)
- Keith Henry Moss
- DTU Health Technology , 202 Kemitorvet , 2800 Kongens Lyngby , Denmark
| | - Petya Popova
- DTU Chemistry , 206-207 Kemitorvet , 2800 Kongens Lyngby , Denmark
| | - Sine R Hadrup
- DTU Health Technology , 202 Kemitorvet , 2800 Kongens Lyngby , Denmark
| | - Kira Astakhova
- DTU Chemistry , 206-207 Kemitorvet , 2800 Kongens Lyngby , Denmark
| | - Maria Taskova
- DTU Chemistry , 206-207 Kemitorvet , 2800 Kongens Lyngby , Denmark
| |
Collapse
|
30
|
Trepotec Z, Lichtenegger E, Plank C, Aneja MK, Rudolph C. Delivery of mRNA Therapeutics for the Treatment of Hepatic Diseases. Mol Ther 2019; 27:794-802. [PMID: 30655211 PMCID: PMC6453508 DOI: 10.1016/j.ymthe.2018.12.012] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/19/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022] Open
Abstract
Promising improvements in the field of transcript therapeutics have clearly enhanced the potential of mRNA as a new pillar for protein replacement therapies. Synthetic mRNAs are engineered to replace mutated mRNAs and to be immunologically inconspicuous and highly stable while maximizing protein expression. Approaches to deliver mRNA into the cellular cytoplasm safely and efficiently have been further developed so that two mRNA-based approaches replacing vascular endothelial growth factor (VEGF) and cystic fibrosis transmembrane conductance regulator (CFTR) have now made it into clinical trials. These studies bring mRNA therapeutics for protein replacement therapy closer to clinical realization. Herein, we provide an overview of preclinical and clinical developments of mRNA therapeutics for liver diseases.
Collapse
Affiliation(s)
- Zeljka Trepotec
- Department of Pediatrics, Ludwig Maximilian University of Munich, 80337 Munich, Germany
| | | | - Christian Plank
- Ethris GmbH, RNA Biology, 82152 Planegg, Germany; Institute of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | | | - Carsten Rudolph
- Department of Pediatrics, Ludwig Maximilian University of Munich, 80337 Munich, Germany; Ethris GmbH, RNA Biology, 82152 Planegg, Germany.
| |
Collapse
|
31
|
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: 712] [Impact Index Per Article: 118.7] [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
|
32
|
Abstract
Immunotherapy has become a powerful clinical strategy for treating cancer. The number of immunotherapy drug approvals has been increasing, with numerous treatments in clinical and preclinical development. However, a key challenge in the broad implementation of immunotherapies for cancer remains the controlled modulation of the immune system, as these therapeutics have serious adverse effects including autoimmunity and nonspecific inflammation. Understanding how to increase the response rates to various classes of immunotherapy is key to improving efficacy and controlling these adverse effects. Advanced biomaterials and drug delivery systems, such as nanoparticles and the use of T cells to deliver therapies, could effectively harness immunotherapies and improve their potency while reducing toxic side effects. Here, we discuss these research advances, as well as the opportunities and challenges for integrating delivery technologies into cancer immunotherapy, and we critically analyse the outlook for these emerging areas.
Collapse
Affiliation(s)
- Rachel S Riley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Patel S, Athirasala A, Menezes PP, Ashwanikumar N, Zou T, Sahay G, Bertassoni LE. Messenger RNA Delivery for Tissue Engineering and Regenerative Medicine Applications. Tissue Eng Part A 2019; 25:91-112. [PMID: 29661055 PMCID: PMC6352544 DOI: 10.1089/ten.tea.2017.0444] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/09/2018] [Indexed: 12/25/2022] Open
Abstract
The ability to control cellular processes and precisely direct cellular reprogramming has revolutionized regenerative medicine. Recent advances in in vitro transcribed (IVT) mRNA technology with chemical modifications have led to development of methods that control spatiotemporal gene expression. Additionally, there is a current thrust toward the development of safe, integration-free approaches to gene therapy for translational purposes. In this review, we describe strategies of synthetic IVT mRNA modifications and nonviral technologies for intracellular delivery. We provide insights into the current tissue engineering approaches that use a hydrogel scaffold with genetic material. Furthermore, we discuss the transformative potential of novel mRNA formulations that when embedded in hydrogels can trigger controlled genetic manipulation to regenerate tissues and organs in vitro and in vivo. The role of mRNA delivery in vascularization, cytoprotection, and Cas9-mediated xenotransplantation is additionally highlighted. Harmonizing mRNA delivery vehicle interactions with polymeric scaffolds can be used to present genetic cues that lead to precise command over cellular reprogramming, differentiation, and secretome activity of stem cells-an ultimate goal for tissue engineering.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
| | - Avathamsa Athirasala
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
| | - Paula P. Menezes
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Postgraduate Program in Health Sciences, Department of Pharmacy, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - N. Ashwanikumar
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
| | - Ting Zou
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, Oregon State University, Portland, Oregon
- Department of Biomedical Engineering, Collaborative Life Science Building, Oregon Health and Science University, Portland, Oregon
| | - Luiz E. Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon
- Department of Biomedical Engineering, Collaborative Life Science Building, Oregon Health and Science University, Portland, Oregon
- Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
34
|
Chen X, Gao C. Influences of size and surface coating of gold nanoparticles on inflammatory activation of macrophages. Colloids Surf B Biointerfaces 2017; 160:372-380. [DOI: 10.1016/j.colsurfb.2017.09.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Accepted: 09/20/2017] [Indexed: 12/31/2022]
|
35
|
Granot Y, Peer D. Delivering the right message: Challenges and opportunities in lipid nanoparticles-mediated modified mRNA therapeutics-An innate immune system standpoint. Semin Immunol 2017; 34:68-77. [PMID: 28890238 DOI: 10.1016/j.smim.2017.08.015] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
mRNA molecules hold tremendous potential as a tool for gene therapy of a wide range of diseases. However, the main hurdle in implementation of mRNA for therapeutics, the systemic delivery of mRNA molecules to target cells, remains a challenge. A feasible solution for this challenge relies in the rapidly evolving field of nucleic acid-loaded nanocarriers and specifically in the established family of lipid-based nanoparticles (LNPs). Herein, we will discuss the main factors, which determine the fate of modified mRNA (mmRNA)-loaded LNPs in-vivo, and will focus on their interactions with the innate immune system as a main consideration in the design of lipid-based mmRNA delivery platforms.
Collapse
Affiliation(s)
- Yasmin Granot
- Laboratory of Precision NanoMedicine, Dept. of Cell Research & Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv 69978, Israel; Dept. of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Dept. of Cell Research & Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv 69978, Israel; Dept. of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
36
|
Abstract
RNA is emerging as a potential therapeutic modality for the treatment of incurable diseases. Despite intense research, the advent to clinical utility remains compromised by numerous biological barriers, hence, there is a need for sophisticated delivery vehicles. In this aspect, lipid nanoparticles (LNPs) are the most advanced platform among nonviral vectors for gene delivery. In this review, we critically review the literature and the reasons for ineffective delivery beyond the liver. We discuss the toxicity issues associated with permanently charged cationic lipids and then turn our attention to next-generation ionizable cationic lipids. These lipids exhibit reduced toxicity and immunogenicity and undergo ionization under the acidic environment of the endosome to release the encapsulated payload to their site of action in the cytosol. Finally, we summarize recent achievements in therapeutic nucleic acid delivery and report on the current status of clinical trials using LNP and the obstacles to clinical translation.
Collapse
Affiliation(s)
- Stephanie Rietwyk
- Laboratory of Precision NanoMedicine, Department of Cell Research & Immunology, George S. Wise Faculty of Life Sciences, ‡Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, §Center for Nanoscience and Nanotechnology, and ∥Cancer Biology Research Center, Tel Aviv University , Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Department of Cell Research & Immunology, George S. Wise Faculty of Life Sciences, ‡Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, §Center for Nanoscience and Nanotechnology, and ∥Cancer Biology Research Center, Tel Aviv University , Tel Aviv 69978, Israel
| |
Collapse
|
37
|
Alvarez MM, Aizenberg J, Analoui M, Andrews AM, Bisker G, Boyden ES, Kamm RD, Karp JM, Mooney DJ, Oklu R, Peer D, Stolzoff M, Strano MS, Trujillo-de Santiago G, Webster TJ, Weiss PS, Khademhosseini A. Emerging Trends in Micro- and Nanoscale Technologies in Medicine: From Basic Discoveries to Translation. ACS NANO 2017; 11:5195-5214. [PMID: 28524668 DOI: 10.1021/acsnano.7b01493] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We discuss the state of the art and innovative micro- and nanoscale technologies that are finding niches and opening up new opportunities in medicine, particularly in diagnostic and therapeutic applications. We take the design of point-of-care applications and the capture of circulating tumor cells as illustrative examples of the integration of micro- and nanotechnologies into solutions of diagnostic challenges. We describe several novel nanotechnologies that enable imaging cellular structures and molecular events. In therapeutics, we describe the utilization of micro- and nanotechnologies in applications including drug delivery, tissue engineering, and pharmaceutical development/testing. In addition, we discuss relevant challenges that micro- and nanotechnologies face in achieving cost-effective and widespread clinical implementation as well as forecasted applications of micro- and nanotechnologies in medicine.
Collapse
Affiliation(s)
- Mario M Alvarez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey , Ave. Eugenio Garza Sada 2501, Col. Tecnológico, CP 64849 Monterrey, Nuevo León, México
| | - Joanna Aizenberg
- Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts 02115, United States
| | - Mostafa Analoui
- UConn Venture Development and Incubation, UConn , Storrs, CT 06269, United States
| | | | | | | | | | | | - David J Mooney
- Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts 02115, United States
| | - Rahmi Oklu
- Division of Interventional Radiology, Mayo Clinic , Scottsdale, Arizona 85259, United States
| | | | | | | | - Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey , Ave. Eugenio Garza Sada 2501, Col. Tecnológico, CP 64849 Monterrey, Nuevo León, México
| | - Thomas J Webster
- Wenzhou Institute of Biomaterials and Engineering, Wenzhou Medical University , Wenzhou 325000, China
| | | | - Ali Khademhosseini
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University , Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| |
Collapse
|
38
|
Controlled and sustained delivery of siRNA/NPs from hydrogels expedites bone fracture healing. Biomaterials 2017; 139:127-138. [PMID: 28601703 DOI: 10.1016/j.biomaterials.2017.06.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/16/2017] [Accepted: 06/02/2017] [Indexed: 01/01/2023]
Abstract
Despite great potential, delivery remains as the most significant barrier to the widespread use of siRNA therapeutics. siRNA has delivery limitations due to susceptibility to RNase degradation, low cellular uptake, and poor tissue-specific localization. Here, we report the development of a hybrid nanoparticle (NP)/hydrogel system that overcomes these challenges. Hydrogels provide localized and sustained delivery via controlled release of entrapped siRNA/NP complexes while NPs protect and enable efficient cytosolic accumulation of siRNA. To demonstrate therapeutic efficacy, regenerative siRNA against WW domain-containing E3 ubiquitin protein ligase 1 (Wwp1) complexed with NP were entrapped within poly(ethylene glycol) (PEG)-based hydrogels and implanted at sites of murine mid-diaphyseal femur fractures. Results showed localization of hydrogels and controlled release of siRNA/NPs at fractures for 28 days, a timeframe over which fracture healing occurs. siRNA/NP sustained delivery from hydrogels resulted in significant Wwp1 silencing at fracture callus compared to untreated controls. Fractures treated with siRNA/NP hydrogels exhibited accelerated bone formation and significantly increased biomechanical strength. This NP/hydrogel siRNA delivery system has outstanding therapeutic promise to augment fracture healing. Owing to the structural similarities of siRNA, the development of the hydrogel platform for in vivo siRNA delivery has myriad therapeutic possibilities in orthopaedics and beyond.
Collapse
|
39
|
Zyuzin MV, Díez P, Goldsmith M, Carregal-Romero S, Teodosio C, Rejman J, Feliu N, Escudero A, Almendral MJ, Linne U, Peer D, Fuentes M, Parak WJ. Comprehensive and Systematic Analysis of the Immunocompatibility of Polyelectrolyte Capsules. Bioconjug Chem 2017; 28:556-564. [DOI: 10.1021/acs.bioconjchem.6b00657] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | - Meir Goldsmith
- Laboratory
of PrecisonNanoMedicine, Department of Cell Research and Immunology,
George S. Wise Faculty of Life Sciences, Department of Materials Science
and Engineering, The Iby and Aladar Fleischman Faculty of Engineering,
Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | | | | | | | - Alberto Escudero
- Instituto
de Ciencia de Materiales de Sevilla, CSIC − Universidad de Sevilla, C. Américo Vespucio 49, E-41092, Seville, Spain
| | - María Jesús Almendral
- Department
of Analytical Chemistry, Nutrition and Food Science, Faculty of Chemistry, University of Salamanca, 37008 Salamanca, Spain
| | | | - Dan Peer
- Laboratory
of PrecisonNanoMedicine, Department of Cell Research and Immunology,
George S. Wise Faculty of Life Sciences, Department of Materials Science
and Engineering, The Iby and Aladar Fleischman Faculty of Engineering,
Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Wolfgang J. Parak
- CIC biomaGUNE, Paseo de Miramón
182, 20014 Donostia
− San Sebastián, Spain
| |
Collapse
|
40
|
Landesman-Milo D, Ramishetti S, Peer D. Nanomedicine as an emerging platform for metastatic lung cancer therapy. Cancer Metastasis Rev 2016; 34:291-301. [PMID: 25948376 DOI: 10.1007/s10555-015-9554-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Metastatic lung cancer is one of the most common cancers leading to mortality worldwide. Current treatment includes chemo- and pathway-dependent therapy aiming at blocking the spread and proliferation of these metastatic lesions. Nanomedicine is an emerging multidisciplinary field that offers unprecedented access to living cells and promises the state of the art in cancer detection and treatment. Development of nanomedicines as drug carriers (nanocarriers) that target cancer for therapy draws upon principles in the fields of chemistry, medicine, physics, biology, and engineering. Given the zealous activity in the field as demonstrated by more than 30 nanocarriers already approved for clinical use and given the promise of recent clinical results in various studies, nanocarrier-based strategies are anticipated to soon have a profound impact on cancer medicine and human health. Herein, we will detail the latest innovations in therapeutic nanomedicine with examples from lipid-based nanoparticles and polymer-based approaches, which are engineered to deliver anticancer drugs to metastatic lung cells. Emphasis will be placed on the latest and most attractive delivery platforms, which are developed specifically to target lung metastatic tumors. These novel nanomedicines may open new avenues for therapeutic intervention carrying new class of drugs such as RNAi and mRNA and the ability to edit the genome using the CRISPER/Cas9 system. Ultimately, these strategies might become a new therapeutic modality for advanced-stage lung cancer.
Collapse
Affiliation(s)
- Dalit Landesman-Milo
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | | | | |
Collapse
|
41
|
Ramishetti S, Landesman-Milo D, Peer D. Advances in RNAi therapeutic delivery to leukocytes using lipid nanoparticles. J Drug Target 2016; 24:780-786. [PMID: 27030014 DOI: 10.3109/1061186x.2016.1172587] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Small interfering RNAs (siRNAs) therapeutics has advanced into clinical trials for liver diseases and solid tumors, but remain a challenge for manipulating leukocytes fate due to lack of specificity and safety issues. Leukocytes ingest pathogens and defend the body through a complex network. They are also involved in the pathogeneses of inflammation, viral infection, autoimmunity and cancers. Modulating gene expression in leukocytes using siRNAs holds great promise to treat leukocyte-mediated diseases. Leukocytes are notoriously hard to transduce with siRNAs and are spread throughout the body often located deep in tissues, therefore developing an efficient systemic delivery strategy is still a challenge. Here, we discuss recent advances in siRNA delivery to leukocyte subsets such as macrophages, monocytes, dendritic cells and lymphocytes. We focus mainly on lipid-based nanoparticles (LNPs) comprised of new generation of ionizable lipids and their ability to deliver siRNA to primary or malignant leukocytes in a targeted manner. Special emphasis is made on LNPs targeted to subsets of leukocytes and we detail a novel microfluidic mixing technology that could aid in changing the landscape of process development of LNPs from a lab tool to a potential novel therapeutic modality.
Collapse
Affiliation(s)
- Srinivas Ramishetti
- a Laboratory of NanoMedicine, Department of Cell Research and Immunology , George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv , Israel.,b Department of Materials Science and Engineering, The Iby and Aladar Fleischman Faculty of Engineering , Tel Aviv University , Tel Aviv , Israel.,c Center for Nanoscience and Nanotechnology , Tel Aviv University , Tel Aviv , Israel
| | - Dalit Landesman-Milo
- a Laboratory of NanoMedicine, Department of Cell Research and Immunology , George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv , Israel.,b Department of Materials Science and Engineering, The Iby and Aladar Fleischman Faculty of Engineering , Tel Aviv University , Tel Aviv , Israel.,c Center for Nanoscience and Nanotechnology , Tel Aviv University , Tel Aviv , Israel
| | - Dan Peer
- a Laboratory of NanoMedicine, Department of Cell Research and Immunology , George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv , Israel.,b Department of Materials Science and Engineering, The Iby and Aladar Fleischman Faculty of Engineering , Tel Aviv University , Tel Aviv , Israel.,c Center for Nanoscience and Nanotechnology , Tel Aviv University , Tel Aviv , Israel
| |
Collapse
|
42
|
Badwaik VD, Aicart E, Mondjinou YA, Johnson MA, Bowman VD, Thompson DH. Structure-property relationship for in vitro siRNA delivery performance of cationic 2-hydroxypropyl-β-cyclodextrin: PEG-PPG-PEG polyrotaxane vectors. Biomaterials 2016; 84:86-98. [PMID: 26826298 PMCID: PMC4755830 DOI: 10.1016/j.biomaterials.2015.11.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/22/2015] [Accepted: 11/25/2015] [Indexed: 11/20/2022]
Abstract
Nanoparticle-mediated siRNA delivery is a promising therapeutic approach, however, the processes required for transport of these materials across the numerous extracellular and intracellular barriers are poorly understood. Efficient delivery of siRNA-containing nanoparticles would ultimately benefit from an improved understanding of how parameters associated with these barriers relate to the physicochemical properties of the nanoparticle vectors. We report the synthesis of three Pluronic(®)-based, cholesterol end-capped cationic polyrotaxanes (PR(+)) threaded with 2-hydroxypropyl-β-cyclodextrin (HPβCD) for siRNA delivery. The biological data showed that PR(+):siRNA complexes were well tolerated (∼90% cell viability) and produced efficient silencing (>80%) in HeLa-GFP and NIH 3T3-GFP cell lines. We further used a multi-parametric approach to identify relationships between the PR(+) structure, PR(+):siRNA complex physical properties, and biological activity. Small angle X-ray scattering and cryoelectron microscopy studies reveal periodicity and lamellar architectures for PR(+):siRNA complexes, whereas the biological assays, ζ potential measurements, and imaging studies suggest that silencing efficiency is influenced by the effective charge ratio (ρeff), polypropylene oxide (PO) block length, and central PO block coverage (i.e., rigidity) of the PR(+) core. We infer from our findings that more compact PR(+):siRNA nanostructures arising from lower molecular weight, rigid rod-like PR(+) polymer cores produce improved silencing efficiency relative to higher molecular weight, more flexible PR(+) vectors of similar effective charge. This study demonstrates that PR(+):siRNA complex formulations can be produced having higher performance than Lipofectamine(®) 2000, while maintaining good cell viability and siRNA sequence protection in cell culture.
Collapse
Affiliation(s)
- Vivek D Badwaik
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Emilio Aicart
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Yawo A Mondjinou
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Merrell A Johnson
- Department of Physics, Indiana University-Purdue University Indianapolis, IN 46202, USA
| | - Valorie D Bowman
- Discovery Park, Hockmeyer Hall of Structural Biology, Purdue University, West Lafayette, IN 47907, USA
| | - David H Thompson
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA.
| |
Collapse
|
43
|
Landesman-Milo D, Peer D. Transforming Nanomedicines From Lab Scale Production to Novel Clinical Modality. Bioconjug Chem 2016; 27:855-62. [PMID: 26734836 DOI: 10.1021/acs.bioconjchem.5b00607] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of nanoparticles as anticancer drug carriers has been studied for over 50 years. These nanoparticles that can carry drugs are now termed "nanomedicines". Since the approval of the first FDA "nanodrug", DOXIL in 1995, tremendous efforts have been made to develop hundreds of nanomedicines based on different materials. The development of drug nanocarriers (NCs) for cancer therapy is especially challenging and requires multidisciplinary approach. Not only is the translation from a lab scale production of the NCs to clinical scale a challenge, but tumor biology and its unique physiology also possess challenges that need to be overcome with cleverer approaches. Yet, with all the efforts made to develop new strategies to deliver drugs (including small molecules and biologics) for cancer therapy, the number of new NCs that are reaching clinical trials is extremely low. Here we discuss the reasons most of the NCs loaded with anticancer drugs are not likely to reach the clinic and emphasize the importance of understanding tumor physiology and heterogeneity, the use of predictive animal models, and the importance of sharing data as key denominators for potential successful translation of NCs from a bench scale into clinical modality for cancer care.
Collapse
Affiliation(s)
- Dalit Landesman-Milo
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University , Tel Aviv 69978, Israel
| |
Collapse
|
44
|
Yung BC, Li J, Zhang M, Cheng X, Li H, Yung EM, Kang C, Cosby LE, Liu Y, Teng L, Lee RJ. Lipid Nanoparticles Composed of Quaternary Amine–Tertiary Amine Cationic Lipid Combination (QTsome) for Therapeutic Delivery of AntimiR-21 for Lung Cancer. Mol Pharm 2016; 13:653-62. [DOI: 10.1021/acs.molpharmaceut.5b00878] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Lesheng Teng
- College of
Life Sciences, Jilin University, Changchun, China
| | - Robert J. Lee
- College of
Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
45
|
Singh MS, Peer D. RNA nanomedicines: the next generation drugs? Curr Opin Biotechnol 2016; 39:28-34. [PMID: 26773301 DOI: 10.1016/j.copbio.2015.12.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/19/2015] [Indexed: 02/08/2023]
Abstract
RNA therapeutics could represent the next generation personalized medicine. The variety of RNA molecules that can inhibit the expression of any mRNA using, for example, RNA interference (RNAi) strategies, or increase the expression of a given protein using modified mRNA together with new gene editing strategies open new avenues for manipulating the fate of diseased cells while leaving healthy cells untouched. In addition, these therapeutic RNA molecules can maximize the treatment of diseases and minimize its adverse effects. Yet, the promise of RNA therapeutics is hindered by the lack of efficient delivery strategies to selectively target these molecules into specific cells. Herein, we will focus on the challenges and opportunities of the delivery of therapeutic RNAi molecules into cancer cells with special emphasis on solid tumors. Solid tumors represent more than 80 percent of cancers and some are very challenging to treat, not merely due to physiological barriers but also since the tumor microenvironment (TME) is a complex milieu of accessory cells besides the cancerous cells. In this review, we will highlight various limiting factors to successful delivery, current clinical achievements and future outlook focusing on RNAi therapeutics to the TME.
Collapse
Affiliation(s)
- Manu Smriti Singh
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Science and Engineering, The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Science and Engineering, The Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
46
|
Moyano DF, Liu Y, Peer D, Rotello VM. Modulation of Immune Response Using Engineered Nanoparticle Surfaces. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:76-82. [PMID: 26618755 PMCID: PMC4749139 DOI: 10.1002/smll.201502273] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/10/2015] [Indexed: 05/28/2023]
Abstract
Nanoparticles (NPs) coated with a monolayer of ligands can be recognized by different components of the immune system, opening new doors for the modulation of immunological responses. By the use of different physical or chemical properties at the NP surface (such as charge, functional groups, and ligand density), NPs can be designed to have distinct cellular uptake, cytokine secretion, and immunogenicity, factors that influence the distribution and clearance of these particles. Understanding these immunological responses is critical for the development of new NP-based carriers for the delivery of therapeutic molecules, and as such several studies have been performed to understand the relationships between immune responses and NP surface functionality. In this review, we will discuss recent reports of these structure-activity relationships, and explore how these motifs can be controlled to elicit therapeutically useful immune responses.
Collapse
Affiliation(s)
- Daniel F. Moyano
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA. Tel: (+1) 413-545-2058
| | - Yuanchang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA. Tel: (+1) 413-545-2058
| | - Dan Peer
- Laboratory of Nanomedicine, Department of Cell Research and Immunology, Department of Materials Science and Engineering, Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv , 69978, Israel. Tel (+972) 3640-7925
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA. Tel: (+1) 413-545-2058
| |
Collapse
|
47
|
Zuckerman JE, Davis ME. Clinical experiences with systemically administered siRNA-based therapeutics in cancer. Nat Rev Drug Discov 2015; 14:843-56. [PMID: 26567702 DOI: 10.1038/nrd4685] [Citation(s) in RCA: 318] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Small interfering RNA (siRNA)-based therapies are emerging as a promising new anticancer approach, and a small number of Phase I clinical trials involving patients with solid tumours have now been completed. Encouraging results from these pioneering clinical studies show that these new therapeutics can successfully and safely inhibit targeted gene products in patients with cancer, and have taught us important lessons regarding appropriate dosages and schedules. In this Review, we critically assess these Phase I studies and discuss their implications for future clinical trial design. Key challenges and future directions in the development of siRNA-containing anticancer therapeutics are also considered.
Collapse
Affiliation(s)
- Jonathan E Zuckerman
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Mark E Davis
- Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
48
|
Abstract
Small interfering RNAs (siRNAs), which downregulate gene expression guided by sequence complementarity, can be used therapeutically to block the synthesis of disease-causing proteins. The main obstacle to siRNA drugs - their delivery into the target cell cytosol - has been overcome to allow suppression of liver gene expression. Here, we review the results of recent clinical trials of siRNA therapeutics, which show efficient and durable gene knockdown in the liver, with signs of promising clinical outcomes and little toxicity. We also discuss the barriers to more widespread applications that target tissues besides the liver and the most promising avenues to overcome them.
Collapse
|
49
|
Zeng H, Johnson ME, Oldenhuis N, Tiambeng TN, Guan Z. Structure-Based Design of Dendritic Peptide Bolaamphiphiles for siRNA Delivery. ACS CENTRAL SCIENCE 2015; 1:303-312. [PMID: 26436138 PMCID: PMC4582325 DOI: 10.1021/acscentsci.5b00233] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Indexed: 05/21/2023]
Abstract
Development of safe and effective delivery vectors is a critical challenge for the application of RNA interference (RNAi)-based biotechnologies. In this study we show the rational design of a series of novel dendritic peptide bolaamphiphile vectors that demonstrate high efficiency for the delivery of small interfering RNA (siRNA) while exhibiting low cytotoxicity and hemolytic activity. Systematic investigation into structure-property relationships revealed an important correlation between molecular design, self-assembled nanostructure, and biological activity. The unique bolaamphiphile architecture proved a key factor for improved complex stability and transfection efficiency. The optimal vector contains a fluorocarbon core and exhibited enhanced delivery efficiency to a variety of cell lines and improved serum resistance when compared to hydrocarbon analogues and lipofectamine RNAiMAX. In addition to introducing a promising new vector system for siRNA delivery, the structure-property relationships and "fluorocarbon effect" revealed herein offer critical insight for further development of novel materials for nucleic acid delivery and other biomaterial applications.
Collapse
|