1
|
Mrksich K, Padilla MS, Mitchell MJ. Breaking the final barrier: Evolution of cationic and ionizable lipid structure in lipid nanoparticles to escape the endosome. Adv Drug Deliv Rev 2024; 214:115446. [PMID: 39293650 DOI: 10.1016/j.addr.2024.115446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/18/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
In the past decade, nucleic acid therapies have seen a boon in development and clinical translation largely due to advances in nanotechnology that have enabled their safe and targeted delivery. Nanoparticles can protect nucleic acids from degradation by serum enzymes and can facilitate entry into cells. Still, achieving endosomal escape to allow nucleic acids to enter the cytoplasm has remained a significant barrier, where less than 5% of nanoparticles within the endo-lysosomal pathway are able to transfer their cargo to the cytosol. Lipid-based drug delivery vehicles, particularly lipid nanoparticles (LNPs), have been optimized to achieve potent endosomal escape, and thus have been the vector of choice in the clinic as demonstrated by their utilization in the COVID-19 mRNA vaccines. The success of LNPs is in large part due to the rational design of lipids that can specifically overcome endosomal barriers. In this review, we chart the evolution of lipid structure from cationic lipids to ionizable lipids, focusing on structure-function relationships, with a focus on how they relate to endosomal escape. Additionally, we examine recent advancements in ionizable lipid structure as well as discuss the future of lipid design.
Collapse
Affiliation(s)
- Kaitlin Mrksich
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marshall S Padilla
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Zhang D, Zhao H, Li P, Wu X, Liang Y. Research Progress on Liposome Pulmonary Delivery of Mycobacterium tuberculosis Nucleic Acid Vaccine and Its Mechanism of Action. J Aerosol Med Pulm Drug Deliv 2024; 37:284-298. [PMID: 38669118 PMCID: PMC11502632 DOI: 10.1089/jamp.2023.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
Traditional vaccines have played an important role in the prevention and treatment of infectious diseases, but they still have problems such as low immunogenicity, poor stability, and difficulty in inducing lasting immune responses. In recent years, the nucleic acid vaccine has emerged as a relatively cheap and safe new vaccine. Compared with traditional vaccines, nucleic acid vaccine has some unique advantages, such as easy production and storage, scalability, and consistency between batches. However, the direct administration of naked nucleic acid vaccine is not ideal, and safer and more effective vaccine delivery systems are needed. With the rapid development of nanocarrier technology, the combination of gene therapy and nanodelivery systems has broadened the therapeutic application of molecular biology and the medical application of biological nanomaterials. Nanoparticles can be used as potential drug-delivery vehicles for the treatment of hereditary and infectious diseases. In addition, due to the advantages of lung immunity, such as rapid onset of action, good efficacy, and reduced adverse reactions, pulmonary delivery of nucleic acid vaccine has become a hot spot in the field of research. In recent years, lipid nanocarriers have become safe, efficient, and ideal materials for vaccine delivery due to their unique physical and chemical properties, which can effectively reduce the toxic side effects of drugs and achieve the effect of slow release and controlled release, and there have been a large number of studies using lipid nanocarriers to efficiently deliver target components into the body. Based on the delivery of tuberculosis (TB) nucleic acid vaccine by lipid carrier, this article systematically reviews the advantages and mechanism of liposomes as a nucleic acid vaccine delivery carrier, so as to lay a solid foundation for the faster and more effective development of new anti-TB vaccine delivery systems in the future.
Collapse
Affiliation(s)
- Danyang Zhang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
- Postgraduate Department of Heibei North University, Zhangjiakou, China
| | - Haimei Zhao
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
- Postgraduate Department of Heibei North University, Zhangjiakou, China
| | - Ping Li
- Postgraduate Department of Heibei North University, Zhangjiakou, China
| | - Xueqiong Wu
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Yan Liang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
3
|
Cong X, Tan H, Lv Y, Mao K, Xin Y, Wang J, Meng X, Guan M, Wang H, Yang YG, Sun T. Impacts of cationic lipid-DNA complexes on immune cells and hematopoietic cells in vivo. Biomater Sci 2024; 12:2381-2393. [PMID: 38500446 DOI: 10.1039/d4bm00148f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
The inability to systemic administration of nanoparticles, particularly cationic nanoparticles, has been a significant barrier to their clinical translation due to toxicity concerns. Understanding the in vivo behavior of cationic lipids is crucial, given their potential impact on critical biological components such as immune cells and hematopoietic stem cells (HSC). These cells are essential for maintaining the body's homeostasis, and their interaction with cationic lipids is a key factor in determining the safety and efficacy of these nanoparticles. In this study, we focused on the cytotoxic effects of cationic lipid/DNA complexes (CLN/DNA). Significantly, we observed that the most substantial cytotoxic effects, including a marked increase in numbers of long-term hematopoietic stem cells (LT-HSC), occurred 24 h post-CLN/DNA treatment in mice. Furthermore, we found that CLN/DNA-induced HSC expansion in bone marrow (BM) led to a notable decrease in the ability to reestablish blood cell production. Our study provides crucial insights into the interaction between cationic lipids and vital cellular components of the immune and hematopoietic systems.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
| | - Huizhu Tan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
| | - Yue Lv
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
- International Center of Future Science, Jilin University, Changchun, Jilin, 130015, China
| | - Yanbao Xin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
| | - Jialiang Wang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
| | - Meng Guan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
| | - Haorui Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
- International Center of Future Science, Jilin University, Changchun, Jilin, 130015, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
- International Center of Future Science, Jilin University, Changchun, Jilin, 130015, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin, 130061, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, 130062, China
- International Center of Future Science, Jilin University, Changchun, Jilin, 130015, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, 130012, China
| |
Collapse
|
4
|
Jia Y, Wang X, Li L, Li F, Zhang J, Liang XJ. Lipid Nanoparticles Optimized for Targeting and Release of Nucleic Acid. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305300. [PMID: 37547955 DOI: 10.1002/adma.202305300] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/25/2023] [Indexed: 08/08/2023]
Abstract
Lipid nanoparticles (LNPs) are currently the most promising clinical nucleic acids drug delivery vehicles. LNPs prevent the degradation of cargo nucleic acids during blood circulation. Upon entry into the cell, specific components of the lipid nanoparticles can promote the endosomal escape of nucleic acids. These are the basic properties of lipid nanoparticles as nucleic acid carriers. As LNPs exhibit hepatic aggregation characteristics, enhancing targeting out of the liver is a crucial way to improve LNPs administrated in vivo. Meanwhile, endosomal escape of nucleic acids loaded in LNPs is often considered inadequate, and therefore, much effort is devoted to enhancing the intracellular release efficiency of nucleic acids. Here, different strategies to efficiently deliver nucleic acid delivery from LNPs are concluded and their mechanisms are investigated. In addition, based on the information on LNPs that are in clinical trials or have completed clinical trials, the issues that are necessary to be approached in the clinical translation of LNPs are discussed, which it is hoped will shed light on the development of LNP nucleic acid drugs.
Collapse
Affiliation(s)
- Yaru Jia
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of HeBei University, Baoding, 071002, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
| | - Xiuguang Wang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of HeBei University, Baoding, 071002, P. R. China
| | - Luwei Li
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of HeBei University, Baoding, 071002, P. R. China
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
| | - Jinchao Zhang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of HeBei University, Baoding, 071002, P. R. China
| | - Xing-Jie Liang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of HeBei University, Baoding, 071002, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
5
|
Khare P, Edgecomb SX, Hamadani CM, E L Tanner E, Manickam DS. Lipid nanoparticle-mediated drug delivery to the brain. Adv Drug Deliv Rev 2023; 197:114861. [PMID: 37150326 DOI: 10.1016/j.addr.2023.114861] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/12/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Lipid nanoparticles (LNPs) have revolutionized the field of drug delivery through their applications in siRNA delivery to the liver (Onpattro) and their use in the Pfizer-BioNTech and Moderna COVID-19 mRNA vaccines. While LNPs have been extensively studied for the delivery of RNA drugs to muscle and liver targets, their potential to deliver drugs to challenging tissue targets such as the brain remains underexplored. Multiple brain disorders currently lack safe and effective therapies and therefore repurposing LNPs could potentially be a game changer for improving drug delivery to cellular targets both at and across the blood-brain barrier (BBB). In this review, we will discuss (1) the rationale and factors involved in optimizing LNPs for brain delivery, (2) ionic liquid-coated LNPs as a potential approach for increasing LNP accumulation in the brain tissue and (3) considerations, open questions and potential opportunities in the development of LNPs for delivery to the brain.
Collapse
Affiliation(s)
- Purva Khare
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA
| | - Sara X Edgecomb
- Department of Chemistry and Biochemistry, The University of Mississippi, MS
| | | | - Eden E L Tanner
- Department of Chemistry and Biochemistry, The University of Mississippi, MS.
| | - Devika S Manickam
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA.
| |
Collapse
|
6
|
Cong X, Tian H, Liu S, Mao K, Chen H, Xin Y, Liu F, Wang X, Meng X, Zhu G, Wang J, Gao X, Tan H, Yang YG, Sun T. Cationic Liposome/DNA Complexes Mediate Antitumor Immunotherapy by Promoting Immunogenic Tumor Cell Death and Dendritic Cell Activation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:28047-28056. [PMID: 32478501 DOI: 10.1021/acsami.0c08112] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Immunotherapy has been successfully used in the treatment of multiple malignancies, but clinical studies revealed low response rates. Thus, the development of new effective immunotherapeutic modalities is urgently needed. Successfully inducing tumor cell death with enhanced antigenicity is important for the expansion and differentiation of tumor-specific CD8+ cytotoxic T lymphocytes. Cationic liposome/DNA complexes (CLN/DNA), which usually have obvious cytotoxic effects, may improve the antitumor immunity through enhancing the immunogenicity of dying tumor cells. Herein, we report that a plasmid DNA-encapsulated cationic lipid nanoparticle formulated with cholesterol, DOTAP, and DSPE-mPEG2000 significantly increases the tumor cell death with high antigenicity in vitro. Furthermore, the cationic liposome/DNA complex (CLN/DNA) treatment promotes the activation of dendritic cells (DCs). We also find that the intratumorally injected CLN/DNA successfully promoted the activation of DCs in the tumor-draining lymph node. Importantly, both local tumor growth and distant tumor formation were significantly inhibited by T cell-dependent antitumor immune responses after intratumoral injection of CLN/DNA. This study presents a simple and effective strategy for improving the cancer immunotherapy.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Huimin Tian
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
| | - Hongmei Chen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
| | - Yanbao Xin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Feiqi Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
| | - Xin Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Xue Gao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Huizhu Tan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
7
|
Pardridge WM. Blood-Brain Barrier and Delivery of Protein and Gene Therapeutics to Brain. Front Aging Neurosci 2020; 11:373. [PMID: 31998120 PMCID: PMC6966240 DOI: 10.3389/fnagi.2019.00373] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/19/2019] [Indexed: 01/02/2023] Open
Abstract
Alzheimer’s disease (AD) and treatment of the brain in aging require the development of new biologic drugs, such as recombinant proteins or gene therapies. Biologics are large molecule therapeutics that do not cross the blood-brain barrier (BBB). BBB drug delivery is the limiting factor in the future development of new therapeutics for the brain. The delivery of recombinant protein or gene medicines to the brain is a binary process: either the brain drug developer re-engineers the biologic with BBB drug delivery technology, or goes forward with brain drug development in the absence of a BBB delivery platform. The presence of BBB delivery technology allows for engineering the therapeutic to enable entry into the brain across the BBB from blood. Brain drug development may still take place in the absence of BBB delivery technology, but with a reliance on approaches that have rarely led to FDA approval, e.g., CSF injection, stem cells, small molecules, and others. CSF injection of drug is the most widely practiced approach to brain delivery that bypasses the BBB. However, drug injection into the CSF results in limited drug penetration to the brain parenchyma, owing to the rapid export of CSF from the brain to blood. A CSF injection of a drug is equivalent to a slow intravenous (IV) infusion of the pharmaceutical. Given the profound effect the existence of the BBB has on brain drug development, future drug or gene development for the brain will be accelerated by future advances in BBB delivery technology in parallel with new drug discovery.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
8
|
Betker JL, Jones D, Childs CR, Helm KM, Terrell K, Nagel MA, Anchordoquy TJ. Nanoparticle uptake by circulating leukocytes: A major barrier to tumor delivery. J Control Release 2018; 286:85-93. [PMID: 30030182 PMCID: PMC6936323 DOI: 10.1016/j.jconrel.2018.07.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/05/2018] [Accepted: 07/16/2018] [Indexed: 12/18/2022]
Abstract
Decades of research into improving drug delivery to tumors has documented uptake of particulate delivery systems by resident macrophages in the lung, liver, and spleen, and correlated short circulation times with reduced tumor accumulation. An implicit assumption in these studies is that nanoparticles present in the blood are available for distribution to the tumor. This study documents significant levels of lipoplex uptake by circulating leukocytes, and its effect on distribution to the tumor and other organs. In agreement with previous studies, PEGylation dramatically extends circulation times and enhances tumor delivery. However, our studies suggest that this relationship is not straightforward, and that particle sequestration by leukocytes can significantly alter biodistribution, especially with non-PEGylated nanoparticle formulations. We conclude that leukocyte uptake should be considered in biodistribution studies, and that delivery to these circulating cells may present opportunities for treating viral infections and leukemia.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dallas Jones
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Christine R Childs
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Karen M Helm
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kristina Terrell
- Flow Cytometry Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Maria A Nagel
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Thomas J Anchordoquy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
9
|
Sharma G, Chopra K, Puri S, Bishnoi M, Rishi P, Kaur IP. Topical delivery of TRPsiRNA-loaded solid lipid nanoparticles confer reduced pain sensation via TRPV1 silencing, in rats. J Drug Target 2017; 26:135-149. [PMID: 28670930 DOI: 10.1080/1061186x.2017.1350857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Present work describes a novel composition for encapsulating TRPsiRNA (TRPV1-targeting siRNA) within lipid-matrix (4:1::glyceryl behnate:stearic acid) of SLNs, using suitably modified cold high-pressure homogenisation technique. Optimisation of the method and composition conducted using calf-thymus DNA (ctDNA), to avoid cost of TRPsiRNA molecules, resulted in small size (d50 = 50-100 nm) and high entrapment (77.22-98.5%). Complete masking of extreme negative charge of both ctDNA (-34.50 mV) and TRPsiRNA (-23.98 mV) upon encapsulation in SLNs without employing cationic components is reported herein for the first time. Diffusion-controlled release (90.17% at 72 h) from a rigid matrix shifted to porous matrix (at 24 h) due to solubilisation of stearic acid at 37 °C. Efficient in vitro (HEK293 T cells) and in vivo transfection and expression established the proof-of-concept. PEG600 as supporting-surfactant and vitrifying agent promoted small size, effective transfection and rupture of endosomal membrane to affect endosomal escape. Physiological efficacy in terms of significant increase (p < .0001) in paw-withdrawal-latency, following topical and intradermal application of TRPsiRNA-loaded SLNs, in rats, exposed to thermal hyperalgesia (145 and 182%, respectively) and capsaicin-induced pain (155 and 182%, respectively) indicate effective silencing of skin TRPV1. Significant decrease in intensity and duration (one-fifth) of capsaicin-induced nocifensive behaviour was also observed. Naked TRPsiRNA, however, did not show any effect.
Collapse
Affiliation(s)
- Gaurav Sharma
- a Department of Pharmaceutics , University Institute of Pharmaceutical Sciences Panjab University , Chandigarh , India
| | - Kanwaljit Chopra
- b Department of Pharmacology , University Institute of Pharmaceutical Sciences Panjab University , Chandigarh , India
| | - Sanjeev Puri
- c Department of Biotechnology , University Institute of Engineering and Technology, Panjab University , Chandigarh , India
| | - Mahendra Bishnoi
- d National Agri-Food Biotechnology Institute (Department of Biotechnology, Government of India) , SAS Nagar Mohali , Punjab , India
| | - Praveen Rishi
- e Department of Microbiology , Basic Medical Sciences Block, Panjab University , Chandigarh , India
| | - Indu P Kaur
- a Department of Pharmaceutics , University Institute of Pharmaceutical Sciences Panjab University , Chandigarh , India
| |
Collapse
|
10
|
Ghatak S, Hascall VC, Markwald RR, Feghali-Bostwick C, Artlett CM, Gooz M, Bogatkevich GS, Atanelishvili I, Silver RM, Wood J, Thannickal VJ, Misra S. Transforming growth factor β1 (TGFβ1)-induced CD44V6-NOX4 signaling in pathogenesis of idiopathic pulmonary fibrosis. J Biol Chem 2017; 292:10490-10519. [PMID: 28389561 DOI: 10.1074/jbc.m116.752469] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 04/06/2017] [Indexed: 01/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive clinical syndrome of fatal outcome. The lack of information about the signaling pathways that sustain fibrosis and the myofibroblast phenotype has prevented the development of targeted therapies for IPF. Our previous study showed that isolated fibrogenic lung fibroblasts have high endogenous levels of the hyaluronan receptor, CD44V6 (CD44 variant containing exon 6), which enhances the TGFβ1 autocrine signaling and induces fibroblasts to transdifferentiate into myofibroblasts. NADPH oxidase 4 (NOX4) enzyme, which catalyzes the reduction of O2 to hydrogen peroxide (H2O2), has been implicated in the cardiac and lung myofibroblast phenotype. However, whether CD44V6 regulates NOX4 to mediate tissue repair and fibrogenesis is not well-defined. The present study assessed the mechanism of how TGF-β-1-induced CD44V6 regulates the NOX4/reactive oxygen species (ROS) signaling that mediates the myofibroblast differentiation. Specifically, we found that NOX4/ROS regulates hyaluronan synthesis and the transcription of CD44V6 via an effect upon AP-1 activity. Further, CD44V6 is part of a positive-feedback loop with TGFβ1/TGFβRI signaling that acts to increase NOX4/ROS production, which is required for myofibroblast differentiation, myofibroblast differentiation, myofibroblast extracellular matrix production, myofibroblast invasion, and myofibroblast contractility. Both NOX4 and CD44v6 are up-regulated in the lungs of mice subjected to experimental lung injury and in cases of human IPF. Genetic (CD44v6 shRNA) or a small molecule inhibitor (CD44v6 peptide) targeting of CD44v6 abrogates fibrogenesis in murine models of lung injury. These studies support a function for CD44V6 in lung fibrosis and offer proof of concept for therapeutic targeting of CD44V6 in lung fibrosis disorders.
Collapse
Affiliation(s)
- Shibnath Ghatak
- From the Department of Regenerative Medicine and Cell Biology,
| | - Vincent C Hascall
- the Department of Biomedical Engineering/ND20, Cleveland Clinic, Cleveland, Ohio 44195
| | | | | | - Carol M Artlett
- the Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129
| | - Monika Gooz
- the College of Pharmacy/Pharmaceutical Biomedical Science, Medical University of South Carolina, Charleston, South Carolina 29425
| | | | - Ilia Atanelishvili
- the Division of Rheumatology and Immunology, Department of Medicine, and
| | - Richard M Silver
- the Division of Rheumatology and Immunology, Department of Medicine, and
| | - Jeanette Wood
- Genkyotex, 16 Chemin des Aulx, CH-1228 Plan-les-Ouates Geneva, Switzerland, and
| | - Victor J Thannickal
- the Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-0006
| | - Suniti Misra
- From the Department of Regenerative Medicine and Cell Biology,
| |
Collapse
|
11
|
Ghatak S, Markwald RR, Hascall VC, Dowling W, Lottes RG, Baatz JE, Beeson G, Beeson CC, Perrella MA, Thannickal VJ, Misra S. Transforming growth factor β1 (TGFβ1) regulates CD44V6 expression and activity through extracellular signal-regulated kinase (ERK)-induced EGR1 in pulmonary fibrogenic fibroblasts. J Biol Chem 2017; 292:10465-10489. [PMID: 28389562 DOI: 10.1074/jbc.m116.752451] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 04/06/2017] [Indexed: 01/06/2023] Open
Abstract
The appearance of myofibroblasts is generally thought to be the underlying cause of the fibrotic changes that underlie idiopathic pulmonary fibrosis. However, the cellular/molecular mechanisms that account for the fibroblast-myofibroblast differentiation/activation in idiopathic pulmonary fibrosis remain poorly understood. We investigated the functional role of hyaluronan receptor CD44V6 (CD44 containing variable exon 6 (v6)) for differentiation of lung fibroblast to myofibroblast phenotype. Increased hyaluronan synthesis and CD44 expression have been detected in numerous fibrotic organs. Previously, we found that the TGFβ1/CD44V6 pathway is important in lung myofibroblast collagen-1 and α-smooth-muscle actin synthesis. Because increased EGR1 (early growth response-1) expression has been shown to appear very early and nearly coincident with the expression of CD44V6 found after TGFβ1 treatment, we investigated the mechanism(s) of regulation of CD44V6 expression in lung fibroblasts by TGFβ1. TGFβ1-mediated CD44V6 up-regulation was initiated through EGR1 via ERK-regulated transcriptional activation. We showed that TGFβ1-induced CD44V6 expression is through EGR1-mediated AP-1 (activator protein-1) activity and that the EGR1- and AP-1-binding sites in the CD44v6 promoter account for its responsiveness to TGFβ1 in lung fibroblasts. We also identified a positive-feedback loop in which ERK/EGR1 signaling promotes CD44V6 splicing and found that CD44V6 then sustains ERK signaling, which is important for AP-1 activity in lung fibroblasts. Furthermore, we identified that HAS2-produced hyaluronan is required for CD44V6 and TGFβRI co-localization and subsequent CD44V6/ERK1/EGR1 signaling. These results demonstrate a novel positive-feedback loop that links the myofibroblast phenotype to TGFβ1-stimulated CD44V6/ERK/EGR1 signaling.
Collapse
Affiliation(s)
- Shibnath Ghatak
- From the Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425,
| | - Roger R Markwald
- From the Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Vincent C Hascall
- the Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195
| | - William Dowling
- From the Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425.,the College of Charleston, Charleston, South Carolina 29424
| | | | | | - Gyada Beeson
- Drug Discovery and Biomedical sciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Craig C Beeson
- Drug Discovery and Biomedical sciences, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Mark A Perrella
- the Division of Pulmonary and Critical Care Medicine, Department of Medicine, and the Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, and
| | - Victor J Thannickal
- the Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-0006
| | - Suniti Misra
- From the Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425,
| |
Collapse
|
12
|
Gerile G, Ganbold T, Li Y, Baigude H. Head group configuration increases the biocompatibility of cationic lipids for nucleic acid delivery. J Mater Chem B 2017. [DOI: 10.1039/c7tb00317j] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Intracellular delivery of genetic material is a potentially powerful therapeutic approach for the treatment of genetic diseases.
Collapse
Affiliation(s)
- Gerile Gerile
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- P. R. China
| | - Tsogzolmaa Ganbold
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- P. R. China
| | - Yizheng Li
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- P. R. China
| | - Huricha Baigude
- School of Chemistry & Chemical Engineering
- Inner Mongolia University
- Hohhot
- P. R. China
| |
Collapse
|
13
|
A Small Indel Mutant Mouse Model of Epidermolytic Palmoplantar Keratoderma and Its Application to Mutant-specific shRNA Therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e299. [PMID: 27003758 PMCID: PMC5014458 DOI: 10.1038/mtna.2016.17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/15/2016] [Indexed: 12/13/2022]
Abstract
Epidermolytic palmoplantar keratoderma (EPPK) is a relatively common autosomal-dominant skin disorder caused by mutations in the keratin 9 gene (KRT9), with few therapeutic options for the affected so far. Here, we report a knock-in transgenic mouse model that carried a small insertion–deletion (indel) mutant of Krt9, c.434delAinsGGCT (p.Tyr144delinsTrpLeu), corresponding to the human mutation KRT9/c.500delAinsGGCT (p.Tyr167delinsTrpLeu), which resulted in a human EPPK-like phenotype in the weight-stress areas of the fore- and hind-paws of both Krt9+/mut and Krt9mut/mut mice. The phenotype confirmed that EPPK is a dominant-negative condition, such that mice heterozygotic for the K9-mutant allele (Krt9+/mut) showed a clear EPPK-like phenotype. Then, we developed a mutant-specific short hairpin RNA (shRNA) therapy for EPPK mice. Mutant-specific shRNAs were systematically identified in vitro using a luciferase reporter gene assay and delivered into Krt9+/mut mice. shRNA-mediated knockdown of mutant protein resulted in almost normal morphology and functions of the skin, whereas the same shRNA had a negligible effect in wild-type K9 mice. Our results suggest that EPPK can be treated by gene therapy, and this has significant implications for future clinical application.
Collapse
|
14
|
Betker JL, Anchordoquy TJ. Relating toxicity to transfection: using sphingosine to maintain prolonged expression in vitro. Mol Pharm 2014; 12:264-73. [PMID: 25418523 PMCID: PMC4291780 DOI: 10.1021/mp500604r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cationic reagents are commonly used to facilitate DNA delivery, and transfection experiments are typically initiated in cell culture where the optimal charge ratio is determined. While transfection rates are often enhanced at higher +/- charge ratios, the cellular toxicity associated with the greater amounts of cationic components at elevated charge ratios is often not considered. In addition, the prolonged effects of cationic lipid uptake on cell viability are not evident in a typical 24-48 h transfection experiment. In this study, we compare the transfection efficiency of cationic lipoplexes to effects on viability of cultured cells in both the short and long term (7 days). Our results indicate that, while minimal toxicity is evident 24 h after exposure to DOTAP-based lipoplexes, cell viability continues to decline and ultimately compromises reporter gene expression at longer times. Substitution of a naturally occurring cationic amphiphile, sphingosine, for DOTAP greatly reduces toxicity and allows high expression to be maintained over prolonged periods.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado , Anschutz Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| | | |
Collapse
|
15
|
Gene delivery potential of biofunctional carbonate apatite nanoparticles in lungs. BIOMED RESEARCH INTERNATIONAL 2014; 2014:646787. [PMID: 25143941 PMCID: PMC4131073 DOI: 10.1155/2014/646787] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/03/2014] [Indexed: 11/30/2022]
Abstract
Existing nonviral gene delivery systems to lungs are inefficient and associated with dose limiting toxicity in mammalian cells. Therefore, carbonate apatite (CO3Ap) nanoparticles were examined as an alternative strategy for effective gene delivery to the lungs. This study aimed to (1) assess the gene delivery efficiency of CO3Ap in vitro and in mouse lungs, (2) evaluate the cytotoxicity effect of CO3Ap/pDNA in vitro, and (3) characterize the CO3Ap/pDNA complex formulations. A significantly high level of reporter gene expression was detected from the lung cell line transfected with CO3Ap/pDNA complex prepared in both serum and serum-free medium. Cytotoxicity analysis revealed that the percentage of the viable cells treated with CO3Ap to be almost similar to the untreated cells. Characterization analyses showed that the CO3Ap/pDNA complexes are in a nanometer range with aggregated spherical structures and tended to be more negatively charged. In the lung of mice, highest level of transgene expression was observed when CO3Ap (8 μL) was complexed with 40 μg of pDNA at day 1 after administration. Although massive reduction of gene expression was seen beyond day 1 post administration, the level of expression remained significant throughout the study period.
Collapse
|
16
|
Durable expression of minicircle DNA-liposome-delivered androgen receptor cDNA in mice with hepatocellular carcinoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:156356. [PMID: 24734226 PMCID: PMC3966419 DOI: 10.1155/2014/156356] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 12/25/2013] [Accepted: 01/21/2014] [Indexed: 11/28/2022]
Abstract
Background. The most common gene-based cancer therapies involve the suppression of oncogenic molecules and enhancement of the expression of tumor-suppressor genes. Studies in noncancer disease animal models have shown that minicircle (MC) DNA vectors are easy to deliver and that the proteins from said MC-carrying DNA vectors are expressed over a long period of time. However, delivery of therapeutic genes via a liposome-mediated, MC DNA complex has never been tested in vascular-rich hepatocellular carcinoma (HCC). Liposome-mediated DNA delivery exhibits high in vivo transfection efficiency and minimal systemic immune response, thereby allowing for repetitive interventions. In this study, we evaluated the efficacy of delivering an MC-liposome vector containing a 3.2 kb androgen receptor (AR; HCC metastasis suppressor) cDNA into Hepatitis B Virus- (HBV-) induced HCC mouse livers. Results. Protein expression and promoter luciferase assays revealed that liposome-encapsulated MC-AR resulted in abundant functional expression of AR protein (100 kD) for up to two weeks. The AR cDNA was also successfully delivered into normal livers and diseased livers, where it was persistently expressed. In both normal livers and livers with tumors, the expression of AR was detectable for up to 60 days. Conclusion. Our results show that an MC/liposome delivery system might improve the efficacy of gene therapy in patients with HCC.
Collapse
|
17
|
Abstract
Nonviral vectors which offer a safer and versatile alternative to viral vectors have been developed to overcome problems caused by viral carriers. However, their transfection efficacy or level of expression is substantially lower than viral vectors. Among various nonviral gene vectors, lipid nanoparticles are an ideal platform for the incorporation of safety and efficacy into a single delivery system. In this chapter, we highlight current lipidic vectors that have been developed for gene therapy of tumors and other diseases. The pharmacokinetic, toxic behaviors and clinic trials of some successful lipids particles are also presented.
Collapse
|
18
|
Lee SJ, Yhee JY, Kim SH, Kwon IC, Kim K. Biocompatible gelatin nanoparticles for tumor-targeted delivery of polymerized siRNA in tumor-bearing mice. J Control Release 2013; 172:358-366. [PMID: 24036198 DOI: 10.1016/j.jconrel.2013.09.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 08/20/2013] [Accepted: 09/02/2013] [Indexed: 11/23/2022]
Abstract
Structural modifications of the siRNA backbone improved its physiochemical properties for incorporating in gene carriers without loss of gene-silencing efficacy. These modifications provide a wider variety of choice of vector systems for siRNA delivery. We developed a tumor-targeted siRNA delivery system using polymerized siRNA (poly-siRNA) and natural polymer gelatin. The polymerized siRNA (poly-siRNA) was prepared through self-polymerization of thiol groups at the 5'-end of sense and anti-sense strands of siRNA and was encapsulated in the self-assembled thiolated gelatin (tGel) nanoparticles (NPs) with chemical cross-linking. The resulting poly-siRNA-tGel (psi-tGel) nanoparticles (average of 145 nm in diameter) protect siRNA molecules from enzymatic degradation, and can be reversibly reduced to release functional siRNA molecules in reductive conditions. The psi-tGel NPs presented efficient siRNA delivery in red fluorescence protein expressing melanoma cells (RFP/B16F10) to down-regulate target gene expression. In addition, the NPs showed low toxicity at a high transfection dose of 125 μg/ml psi-tGel NPs, which included 1 μM of siRNA molecules. In tumor-bearing mice, the psi-tGel NPs showed 2.8 times higher tumor accumulation than the naked poly-siRNA, suggesting tumor-targeted siRNA delivery of psi-tGel NPs. Importantly, the psi-tGel NPs induced effective tumor RFP gene silencing in vivo without remarkable toxicity. The psi-tGel NPs have great potential for a systemic siRNA delivery system for cancer therapy, based on their characteristics of low toxicity, tumor accumulation, and effective siRNA delivery.
Collapse
Affiliation(s)
- So Jin Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| | - Ji Young Yhee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea; KU-KIST School, Korea University, 1 Anam-dong, Seongbuk-gu, Seoul 136-701, South Korea.
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 6, Seongbuk-gu, Seoul 136-791, South Korea.
| |
Collapse
|
19
|
Kaneda Y. Virosome: a novel vector to enable multi-modal strategies for cancer therapy. Adv Drug Deliv Rev 2012; 64:730-8. [PMID: 21443915 DOI: 10.1016/j.addr.2011.03.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 02/22/2011] [Accepted: 03/20/2011] [Indexed: 01/11/2023]
Abstract
Despite advancements in treatments, cancer remains a life-threatening disease that is resistant to therapy. Single-modal cancer therapy is often insufficient to provide complete remission. A revolution in cancer therapy may someday be provided by vector-based gene and drug delivery systems. However, it remains difficult to achieve this aim because viral and non-viral vectors have their own advantages and limitations. To overcome these limitations, virosomes have been constructed by combining viral components with non-viral vectors or by using pseudovirions without viral genome replication. Viruses, such as influenza virus, HVJ (hemagglutinating virus of Japan; Sendai virus) and hepatitis B virus, have been used in the construction of virosomes. The HVJ-derived vector is particularly promising due to its highly efficient delivery of DNA, siRNA, proteins and anti-cancer drugs. Furthermore, the HVJ envelope (HVJ-E) vector has intrinsic anti-tumor activities including the activation of multiple anti-tumor immunities and the induction of cancer-selective apoptosis. HVJ-E is currently being clinically used for the treatment of melanoma. A promising multi-modal cancer therapy will be achieved when virosomes with intrinsic anti-tumor activities are utilized as vectors for the delivery of anti-tumor drugs and genes.
Collapse
Affiliation(s)
- Yasufumi Kaneda
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, Suita, Japan.
| |
Collapse
|
20
|
Ansaldi D, Hod EA, Stellari F, Kim JB, Lim E, Roskey M, Francis KP, Singh R, Zhang N. Imaging pulmonary NF-kappaB activation and therapeutic effects of MLN120B and TDZD-8. PLoS One 2011; 6:e25093. [PMID: 21966423 PMCID: PMC3178604 DOI: 10.1371/journal.pone.0025093] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 08/24/2011] [Indexed: 12/14/2022] Open
Abstract
NF-κB activation is a critical signaling event in the inflammatory response and has been implicated in a number of pathological lung diseases. To enable the assessment of NF-κB activity in the lungs, we transfected a luciferase based NF-κB reporter into the lungs of mice or into Raw264.7 cells in culture. The transfected mice showed specific luciferase expression in the pulmonary tissues. Using these mouse models, we studied the kinetics of NF-κB activation following exposure to lipopolysaccharide (LPS). The Raw264.7 cells expressed a dose-dependent increase in luciferase following exposure to LPS and the NF-κB reporter mice expressed luciferase in the lungs following LPS challenge, establishing that bioluminescence imaging provides adequate sensitivity for tracking the NF-κB activation pathway. Interventions affecting the NF-κB pathway are promising clinical therapeutics, thus we further examined the effect of IKK-2 inhibition by MLN120B and glycogen synthase kinase 3 beta inhibition by TDZD-8 on NF-κB activation. Pre-treatment with either MLN120B or TDZD-8 attenuated NF-κB activation in the pulmonary tissues, which was accompanied with suppression of pro-inflammatory chemokine MIP-1ß and induction of anti-inflammatory cytokine IL-10. In summary, we have established an imaging based approach for non-invasive and longitudinal assessment of NF-κB activation and regulation during acute lung injury. This approach will potentiate further studies on NF-κB regulation under various inflammatory conditions.
Collapse
Affiliation(s)
- Dan Ansaldi
- Caliper Life Sciences, Alameda, California, United States of America
| | - Eldad A. Hod
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - Fabio Stellari
- In Vivo Pulmonary Pharmacology Department, Chiesi Group, Parma, Italy
| | - Jae-Beom Kim
- Caliper Life Sciences, Alameda, California, United States of America
| | - Ed Lim
- Caliper Life Sciences, Alameda, California, United States of America
| | - Mark Roskey
- Caliper Life Sciences, Alameda, California, United States of America
| | - Kevin P. Francis
- Caliper Life Sciences, Alameda, California, United States of America
| | - Rajendra Singh
- Caliper Life Sciences, Alameda, California, United States of America
| | - Ning Zhang
- Caliper Life Sciences, Alameda, California, United States of America
- * E-mail:
| |
Collapse
|
21
|
Kaneda Y. Update on non-viral delivery methods for cancer therapy: possibilities of a drug delivery system with anticancer activities beyond delivery as a new therapeutic tool. Expert Opin Drug Deliv 2010; 7:1079-93. [PMID: 20716020 DOI: 10.1517/17425247.2010.510511] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Cancer is the most formidable human disease. Owing to the heterogeneity of cancer, a single-treatment modality is insufficient for the complete elimination of cancer cells. Therapeutic strategies from various aspects are needed for cancer therapy. These therapeutic agents should be carefully selected to enhance multiple therapeutic pathways. Non-viral delivery methods have been utilized to enhance the tumor-selective delivery of therapeutic molecules, including proteins, synthetic oligonucleotides, small compounds and genes. AREAS COVERED IN THIS REVIEW As non-viral delivery methods, liposomes and polymer-based delivery materials to target tumors mainly by systemic delivery, physical methods including electroporation, sonoporation, and so on, to locally inject therapeutic molecules, and virosomes to use the viral infectious machinery for the delivery of therapeutic molecules are summarized. WHAT THE READER WILL GAIN This article aims to provide an overview of the characteristic properties of each non-viral vector. It will be beneficial to utilize appropriately the vector for cancer treatment. TAKE HOME MESSAGE Efficient and minimally invasive vectors are generally considered to be the ideal drug delivery system (DDS). However, against cancer, DDS equipped with antitumor activities may be a therapeutic choice. By combining therapeutic molecules with DDS having antitumor activities, enhancement of the multiple therapeutic pathways may be achieved.
Collapse
Affiliation(s)
- Yasufumi Kaneda
- Osaka University, Graduate School of Medicine, Division of Gene Therapy Science, Suita, Osaka, Japan.
| |
Collapse
|
22
|
Zimmerman AL, Wu S. MicroRNAs, cancer and cancer stem cells. Cancer Lett 2010; 300:10-9. [PMID: 20965651 DOI: 10.1016/j.canlet.2010.09.019] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 09/27/2010] [Accepted: 09/28/2010] [Indexed: 12/19/2022]
Abstract
MicroRNAs regulate self-renewal, differentiation, and division of cells via post-transcriptional gene silencing. Aberrant microRNA levels, specifically an overall downregulation, are present in many cancers, as compared to their normal tissue counterparts. Therefore, a potential therapeutic use of microRNAs is to correct these aberrant transcript levels involved in the signaling pathways of cancer. This review focuses on the current knowledge of microRNAs and their involvement with cancer cells and cancer stem cells. The methods currently being used to develop miRNA-based cancer therapeutics are examined, and the limitations halting further progress are also discussed.
Collapse
Affiliation(s)
- Amy L Zimmerman
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, United States; Honors Tutorial College, Ohio University, Athens, OH 45701, United States
| | | |
Collapse
|
23
|
Comparative assessment of siRNA and shRNA off target effects: what is slowing clinical development. Cancer Gene Ther 2009; 16:807-9. [DOI: 10.1038/cgt.2009.53] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
24
|
Lacaze P, Raza S, Sing G, Page D, Forster T, Storm P, Craigon M, Awad T, Ghazal P, Freeman TC. Combined genome-wide expression profiling and targeted RNA interference in primary mouse macrophages reveals perturbation of transcriptional networks associated with interferon signalling. BMC Genomics 2009; 10:372. [PMID: 19664281 PMCID: PMC2741489 DOI: 10.1186/1471-2164-10-372] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Accepted: 08/10/2009] [Indexed: 01/09/2023] Open
Abstract
Background Interferons (IFNs) are potent antiviral cytokines capable of reprogramming the macrophage phenotype through the induction of interferon-stimulated genes (ISGs). Here we have used targeted RNA interference to suppress the expression of a number of key genes associated with IFN signalling in murine macrophages prior to stimulation with interferon-gamma. Genome-wide changes in transcript abundance caused by siRNA activity were measured using exon-level microarrays in the presence or absence of IFNγ. Results Transfection of murine bone-marrow derived macrophages (BMDMs) with a non-targeting (control) siRNA and 11 sequence-specific siRNAs was performed using a cationic lipid transfection reagent (Lipofectamine2000) prior to stimulation with IFNγ. Total RNA was harvested from cells and gene expression measured on Affymetrix GeneChip Mouse Exon 1.0 ST Arrays. Network-based analysis of these data revealed six siRNAs to cause a marked shift in the macrophage transcriptome in the presence or absence IFNγ. These six siRNAs targeted the Ifnb1, Irf3, Irf5, Stat1, Stat2 and Nfkb2 transcripts. The perturbation of the transcriptome by the six siRNAs was highly similar in each case and affected the expression of over 600 downstream transcripts. Regulated transcripts were clustered based on co-expression into five major groups corresponding to transcriptional networks associated with the type I and II IFN response, cell cycle regulation, and NF-KB signalling. In addition we have observed a significant non-specific immune stimulation of cells transfected with siRNA using Lipofectamine2000, suggesting use of this reagent in BMDMs, even at low concentrations, is enough to induce a type I IFN response. Conclusion Our results provide evidence that the type I IFN response in murine BMDMs is dependent on Ifnb1, Irf3, Irf5, Stat1, Stat2 and Nfkb2, and that siRNAs targeted to these genes results in perturbation of key transcriptional networks associated with type I and type II IFN signalling and a suppression of macrophage M1 polarization.
Collapse
Affiliation(s)
- Paul Lacaze
- Division of Pathway Medicine, The University of Edinburgh, The Chancellor's Building, College of Medicine, Edinburgh, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chikh G, de Jong SD, Sekirov L, Raney SG, Kazem M, Wilson KD, Cullis PR, Dutz JP, Tam YK. Synthetic methylated CpG ODNs are potent in vivo adjuvants when delivered in liposomal nanoparticles. Int Immunol 2009; 21:757-67. [PMID: 19502586 DOI: 10.1093/intimm/dxp044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although it is well documented that the immunological activity of cytosine-guanine (CpG) motifs is abrogated by 5' methylation of the cytosine residue, encapsulation within stabilized lipid nanoparticles endows these methylated cytosine-guanine- (mCpG-) containing oligonucleotides (ODNs) with potent immunostimulatory activity in murine animal models. Surprisingly, not only do liposomal nanoparticulate (LN) mCpG ODN possess immunostimulatory activity, their potency is found to be equivalent and often greater than the equivalent unmethylated form, as judged by a number of ex vivo innate and adaptive immune parameters and anti-tumor efficacy in murine models. Preliminary data indicate that both methylated and unmethylated CpG ODN act through a common receptor signaling pathway, specifically via toll-like receptor (TLR) 9, based on observations of up-regulated TLR9 expression, induction of nitric oxide and dependence on endosomal maturation. This is confirmed in TLR9 knockout animals which show no immunostimulatory activity following treatment with LN-mCpG ODN. These data, therefore, indicate that the mCpG DNA is fully competent to interact with TLR9 to initiate potent immune responses. Furthermore, this work implicates an as yet unidentified mechanism upstream of TLR9 which regulates the relative activities of free methylated versus unmethylated CpG ODN that is effectively bypassed by particulate delivery of CpG ODN.
Collapse
Affiliation(s)
- Ghania Chikh
- Tekmira Pharmaceuticals Corporation, British Columbia, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Burckbuchler V, Wintgens V, Leborgne C, Lecomte S, Leygue N, Scherman D, Kichler A, Amiel C. Development and Characterization of New Cyclodextrin Polymer-Based DNA Delivery Systems. Bioconjug Chem 2008; 19:2311-20. [DOI: 10.1021/bc800070f] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | | | | | | | - Catherine Amiel
- Institut de Chimie des Matériaux Paris Est, Systèmes Polymères Complexes, Université Paris 12 Val de Marne, CNRS, 2-8 rue Henri Dunant, 94320 Thiais, France, FRE3087 CNRS-Généthon, 1 rue de l’Internationale, BP 60, 91002 Evry Cedex, France, and Laboratoire de Dynamique, Interactions et Réactivité, CNRS, 2-8 rue Henri Dunant, 94320 Thiais, France
| |
Collapse
|
27
|
Zhao J, Wen Y, Li Q, Wang Y, Wu H, Xu J, Chen X, Wu Y, Fan L, Yang H, Liu T, Ding Z, Du X, Diao P, Li J, Wu H, Kan B, Lei S, Deng H, Mao Y, Zhao X, Wei Y. A promising cancer gene therapy agent based on the matrix protein of vesicular stomatitis virus. FASEB J 2008; 22:4272-80. [PMID: 18716030 DOI: 10.1096/fj.08-110049] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ju‐mei Zhao
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
- Pharmacological Department of Medical CollegeYan'an UniversityYan'anshanxiChina
| | - Yan‐jun Wen
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Qiu Li
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Yong‐sheng Wang
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Hong‐bo Wu
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Jian‐rong Xu
- College of Life ScienceSichuan UniversityChengduSichuanChina
| | - Xian‐cheng Chen
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Yang Wu
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Ling‐yu Fan
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Han‐shuo Yang
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Tao Liu
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Zhen‐yu Ding
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Xiao‐bo Du
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Peng Diao
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Jiong Li
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Hong‐bing Wu
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Bing Kan
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Song Lei
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Hong‐xin Deng
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Yong‐qiu Mao
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Xia Zhao
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| | - Yu‐quan Wei
- State Key Laboratory of BiotherapyWest China Hospital, and Sichuan UniversityChengduSichuanChina
| |
Collapse
|
28
|
|
29
|
Liu K, Anderson GP, Bozinovski S. DNA vector augments inflammation in epithelial cells via EGFR-dependent regulation of TLR4 and TLR2. Am J Respir Cell Mol Biol 2008; 39:305-11. [PMID: 18403779 DOI: 10.1165/rcmb.2007-0458oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gene delivery applications to treat lung diseases are, in some instances, suboptimal due to deleterious host inflammatory reactions. Current DNA plasmids (pDNA) exert toxicity in part via unmethylated CpG motifs that stimulate Toll-like receptor (TLR)9-expressing leukocytes; however, the airway epithelial response has not been well defined. Bronchial epithelial cells (BEAS-2B) were exposed to pDNA complexes and inflammatory mediators were measured. As patients with inflammatory lung disease are susceptible to infectious exacerbations, we also evaluated the reciprocal inflammatory response to pDNA and bacterial components lipopolysaccharide (LPS) and lipoteichoic acid (LTA), recognized by TLR4 and TLR2, respectively. Cells primed with pDNA synergistically expressed IL-8 mRNA and protein in response to LPS and LTA (3- to 5-fold). A similar induction was also observed for IL-1beta, IL-6, colony-stimulating factor (CSF)-1, and granulocyte macrophage-CSF. Their synergistic elevation was associated with an increase in TLR4 and TLR2 levels. Methylation of pDNA only partially reduced (25-30%) IL-8 release; hence, signaling occurs via CpG/TLR9-dependent and -independent modules. As epidermal growth factor receptor (EGFR) signaling has been implicated in bronchial IL-8 expression, we assessed whether pDNA priming events were coordinated via EGFR. AG1478 (EGFR inhibitor) restored normal TLR4/2 levels and also suppressed synergistic release of IL-8. The extracellular signal-regulated kinase (Erk) mitogen-activated protein kinase inhibitor also blocked IL-8 release, implicating Erk as a key mediator of EGFR signaling. Our findings identify a novel EGFR-dependent mechanism for regulating TLR, and show that targeted disruption of EGFR signaling ameliorates the airway epithelial inflammatory response to pDNA. Targeting the EGFR system may improve the efficiency, tolerability, and safety of gene therapy strategies.
Collapse
Affiliation(s)
- Kenneth Liu
- Department of Pharmacology, The University of Melbourne, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
30
|
Chen W, Yan W, Huang L. A simple but effective cancer vaccine consisting of an antigen and a cationic lipid. Cancer Immunol Immunother 2008; 57:517-30. [PMID: 17724588 PMCID: PMC11029866 DOI: 10.1007/s00262-007-0390-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Accepted: 08/10/2007] [Indexed: 12/11/2022]
Abstract
Developing a cancer vaccine with a potent adjuvant, which is safe for human use, remains to be an unmet need. In this study, we developed a simple, safe, yet efficient, peptide-based therapeutic cancer vaccine, DOTAP/E7 complex, which comprises only two molecules: a DOTAP cationic lipid and a peptide antigen derived from E7 oncoprotein of human papillomavirus (HPV) type 16. The anti-cancer activity of DOTAP/E7 against existing HPV positive TC-1 tumor was compared to that of our previous LPD/E7 formulation, which contains bacterial DNA CpG motifs. Tumor-bearing mice showed significant tumor inhibition following a single vaccination of either formulation at the optimal lipid dose, suggesting that DOTAP liposome alone can provide a potent adjuvant activity without plasmid DNA. E7 peptide formulated with DOTAP induced migration of activated dendritic cells (DC) to the draining lymph node (DLN) and efficiently generated functional antigen-specific CD8+ T lymphocyte responses. Accumulation of CD8+ tumor infiltrating T cells and apoptosis at tumor sites were observed after treatment with DOTAP/E7 complexes, which was also associated with a decreased amount of CD25(+)Foxp3(+) regulatory T cells in treated animals. Reactive oxygen species (ROS) induced by DOTAP cationic lipid in DLN revealed a plausible mechanism of the initial interaction between DC and DOTAP. An adequate amount of ROS generation was apparently required for the initiation of the vaccine mechanism; however, an overdose of DOTAP induced massive ROS production and apoptosis of DC in DLN, which led to diminished anti-cancer immunity. Overall, these results indicate that cationic lipid DOTAP alone serves as an efficient vaccine adjuvant for the induction of a therapeutic, antigen-specific anti-cancer activity.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cations/immunology
- Cytotoxicity, Immunologic
- Fatty Acids, Monounsaturated/immunology
- Fatty Acids, Monounsaturated/metabolism
- Female
- Flow Cytometry
- Immunohistochemistry
- In Situ Nick-End Labeling
- Lipids/immunology
- Liposomes/immunology
- Mice
- Mice, Inbred C57BL
- Neoplasms, Experimental/therapy
- Oncogene Proteins, Viral/immunology
- Oncogene Proteins, Viral/metabolism
- Papillomavirus E7 Proteins
- Quaternary Ammonium Compounds/immunology
- Quaternary Ammonium Compounds/metabolism
- Reactive Oxygen Species
Collapse
Affiliation(s)
- Weihsu Chen
- Division of Molecular Pharmaceutics, School of Pharmacy, University of North Carolina at Chapel Hill, 2316 Kerr Hall, CB# 7360, Chapel Hill, NC 27599 USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15260 USA
| | - Weili Yan
- Division of Molecular Pharmaceutics, School of Pharmacy, University of North Carolina at Chapel Hill, 2316 Kerr Hall, CB# 7360, Chapel Hill, NC 27599 USA
| | - Leaf Huang
- Division of Molecular Pharmaceutics, School of Pharmacy, University of North Carolina at Chapel Hill, 2316 Kerr Hall, CB# 7360, Chapel Hill, NC 27599 USA
| |
Collapse
|
31
|
Chen ZY, Riu E, He CY, Xu H, Kay MA. Silencing of episomal transgene expression in liver by plasmid bacterial backbone DNA is independent of CpG methylation. Mol Ther 2008; 16:548-56. [PMID: 18253155 DOI: 10.1038/sj.mt.6300399] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Minicircle DNA vectors devoid of plasmid bacterial backbone, (BB) DNAs, are transcriptionally persistent, whereas their parent plasmid counterparts are silenced in the liver. In this study we establish that circular plasmid BB provided in trans did not silence a transgene expression cassette in vivo, further confirming our previous conclusions that the covalent attachment of the plasmid BB to the expression cassette is required for DNA silencing. Given the high concentration of CpG dinucleotides in the plasmid BB, we investigated the role of DNA methylation on transgene silencing in vivo. The presence or absence of methylation in CpG motifs in routine plasmid BBs had no significant effect on transcriptional silencing. Furthermore, the removal of the CpG motifs from the BB did not ameliorate transcriptional silencing. Transgene silencing was partially inhibited when two tandem copies of the chicken cHS4 insulator at each end of a routine plasmid vector were used. These results are consistent with the idea that the transcriptional repression observed with plasmid DNA vectors in the liver is caused by formation of repressive heterochromatin on the plasmid DNA backbone, which then spreads and inactivates the transgene in cis, and that CpG content or methylation has little or no influence in the process.
Collapse
Affiliation(s)
- Zhi-Ying Chen
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
Gene delivery using nonviral approaches has been extensively studied as a basic tool for intracellular gene transfer and gene therapy. In the past, the primary focus has been on application of physical, chemical, and biological principles to development of a safe and efficient method that delivers a transgene into target cells for appropriate expression. This review summarizes the current status of the most commonly used nonviral methods, with an emphasis on their mechanism of action for gene delivery, and their advantages and limitations for gene therapy applications. The technical aspects of each delivery system are also reviewed, with a focus on how to achieve optimal delivery efficiency. A brief discussion of future development and further improvement of the current systems is intended to stimulate new ideas and encourage rapid advancement in this new and promising field.
Collapse
Affiliation(s)
- Xiang Gao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 15261 Pittsburgh, PA
| | - Keun-Sik Kim
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 15261 Pittsburgh, PA
| | - Dexi Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 15261 Pittsburgh, PA
| |
Collapse
|
33
|
Zhou R, Norton JE, Zhang N, Dean DA. Electroporation-mediated transfer of plasmids to the lung results in reduced TLR9 signaling and inflammation. Gene Ther 2007; 14:775-80. [PMID: 17344904 PMCID: PMC4150868 DOI: 10.1038/sj.gt.3302936] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Electroporation can deliver DNA efficiently and safely to tissues in live animals, including the lung where it causes little inflammation or lung injury. In contrast, cationic lipid-mediated gene transfer has been shown to induce an inflammatory response caused by unmethylated plasmid CpG residues, which activate the toll-like receptor (TLR9) signaling pathway. As TLR9 is located in the endosomal/lysosomal compartment, we hypothesized that plasmids do not activate TLR9 during electroporation because they enter the cytoplasm directly through transient pores in the plasma membrane. To test this, plasmids were transfected into kidney epithelial cells overexpressing TLR9 (HEK293-TLR9+) and cells lacking TLR9 (HEK293-TLR9-null). Interleukin (IL)-8 expression, an indicator of TLR9 activation, increased more than 10-fold at 24 h post-liposome transfection in HEK293-TLR9+ cells, but showed no significant increase in electroporated cells, compared with untransfected cells. In vivo liposome-mediated gene transfer caused increases in IL-6, IL-12, tumor necrosis factor alpha and interferon gamma in mouse bronchial alveolar lavage fluid, whereas the levels of these cytokines were more than 10-fold lower by comparison following electroporation. Depletion of alveolar macrophages suggested that this inflammatory response is mediated by resident pulmonary epithelial cells. These results suggest that electroporation-mediated gene transfer bypasses the TLR-9 pathway, thus accounting for the low levels of inflammation seen with this approach.
Collapse
Affiliation(s)
- Rui Zhou
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - James E. Norton
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Ning Zhang
- Division of Allergy-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - David A. Dean
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Address all correspondence to: David A. Dean, Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, 240 E. Huron Ave, McGaw 2336, Chicago, IL 60611, Tel: 312-503-3121, Fax: 312-908-4650,
| |
Collapse
|
34
|
Sakurai H, Sakurai F, Kawabata K, Sasaki T, Koizumi N, Huang H, Tashiro K, Kurachi S, Nakagawa S, Mizuguchi H. Comparison of gene expression efficiency and innate immune response induced by Ad vector and lipoplex. J Control Release 2006; 117:430-7. [PMID: 17239467 DOI: 10.1016/j.jconrel.2006.11.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2006] [Revised: 10/27/2006] [Accepted: 11/28/2006] [Indexed: 12/30/2022]
Abstract
Vectors for gene expression are the essential tools for both gene therapy and basic research. There are two groups of gene therapy vectors, viral and non-viral vectors. At present, toxicity triggered by vectors is one of the major concerns for clinical trials. In general, non-viral vectors, such as plasmid DNA-cationic liposome complex (lipoplex), are thought to be safer than viral vectors, such as adenovirus (Ad) vector, although lipoplex is less efficient in term of gene expression than the Ad vector. However, there has been no study directly comparing the gene expression efficiency and safety of viral and non-viral vectors. Here, we present evidence that the Ad vector shows much more efficient gene expression and is safer than lipoplex, at least with respect to the innate immune response. After being systemically administered to mice, the Ad vector showed a transduction efficiency that was 2 to 5 log orders higher than that of lipoplex, depending on the organ. On the other hand, surprisingly, the administration of lipoplex produced a greater amount of inflammatory cytokines such as interleukin-6, interleukin-12, and tumor necrosis factor-alpha than did the administration of the Ad vector, whereas a comparable level of hepatotoxicity was induced by these vectors. The production of inflammatory cytokines induced by the injection of lipoplex was reduced when the CpG motifs were removed completely from plasmid DNA. Thus, care should be taken to ensure the innate immune response induced by gene therapy vectors, especially lipoplex.
Collapse
Affiliation(s)
- Haruna Sakurai
- Laboratory of Gene Transfer and Regulation, National Institute of Biomedical Innovation, Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
|