1
|
Leenaars CHC, Stafleu FR, Häger C, Bleich A. A case study of the informative value of risk of bias and reporting quality assessments for systematic reviews. Syst Rev 2024; 13:230. [PMID: 39244603 PMCID: PMC11380326 DOI: 10.1186/s13643-024-02650-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024] Open
Abstract
While undisputedly important, and part of any systematic review (SR) by definition, evaluation of the risk of bias within the included studies is one of the most time-consuming parts of performing an SR. In this paper, we describe a case study comprising an extensive analysis of risk of bias (RoB) and reporting quality (RQ) assessment from a previously published review (CRD42021236047). It included both animal and human studies, and the included studies compared baseline diseased subjects with controls, assessed the effects of investigational treatments, or both. We compared RoB and RQ between the different types of included primary studies. We also assessed the "informative value" of each of the separate elements for meta-researchers, based on the notion that variation in reporting may be more interesting for the meta-researcher than consistently high/low or reported/non-reported scores. In general, reporting of experimental details was low. This resulted in frequent unclear risk-of-bias scores. We observed this both for animal and for human studies and both for disease-control comparisons and investigations of experimental treatments. Plots and explorative chi-square tests showed that reporting was slightly better for human studies of investigational treatments than for the other study types. With the evidence reported as is, risk-of-bias assessments for systematic reviews have low informative value other than repeatedly showing that reporting of experimental details needs to improve in all kinds of in vivo research. Particularly for reviews that do not directly inform treatment decisions, it could be efficient to perform a thorough but partial assessment of the quality of the included studies, either of a random subset of the included publications or of a subset of relatively informative elements, comprising, e.g. ethics evaluation, conflicts of interest statements, study limitations, baseline characteristics, and the unit of analysis. This publication suggests several potential procedures.
Collapse
Affiliation(s)
- Cathalijn H C Leenaars
- Institute for Laboratory Animal Science, Hannover Medical School, Carl Neubergstrasse 1, 30625, Hannover, Germany.
| | - Frans R Stafleu
- Department of Animals in Science and Society, Utrecht University, Yalelaan 2, Utrecht, 3584 CM, the Netherlands
| | - Christine Häger
- Institute for Laboratory Animal Science, Hannover Medical School, Carl Neubergstrasse 1, 30625, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Carl Neubergstrasse 1, 30625, Hannover, Germany
| |
Collapse
|
2
|
Waters S, Middleton PG. Rectal organoid morphometric analysis (ROMA)-re: optimising measurements automatically. Thorax 2024; 79:801-802. [PMID: 39025505 DOI: 10.1136/thorax-2024-221829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 07/20/2024]
Affiliation(s)
| | - Peter G Middleton
- Respiratory & Sleep Medicine, Westmead Hospital, Westmead, New South Wales, Australia
- Westmead Clinical School, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Vlashi R, Zhang X, Li H, Chen G. Potential therapeutic strategies for osteoarthritis via CRISPR/Cas9 mediated gene editing. Rev Endocr Metab Disord 2024; 25:339-367. [PMID: 38055160 DOI: 10.1007/s11154-023-09860-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
Osteoarthritis (OA) is an incapacitating and one of the most common physically degenerative conditions with an assorted etiology and a highly complicated molecular mechanism that to date lacks an efficient treatment. The capacity to design biological networks and accurately modify existing genomic sites holds an apt potential for applications across medical and biotechnological sciences. One of these highly specific genomes editing technologies is the CRISPR/Cas9 mechanism, referred to as the clustered regularly interspaced short palindromic repeats, which is a defense mechanism constituted by CRISPR associated protein 9 (Cas9) directed by small non-coding RNAs (sncRNA) that bind to target DNA through Watson-Crick base pairing rules where subsequent repair of the target DNA is initiated. Up-to-date research has established the effectiveness of the CRISPR/Cas9 mechanism in targeting the genetic and epigenetic alterations in OA by suppressing or deleting gene expressions and eventually distributing distinctive anti-arthritic properties in both in vitro and in vivo osteoarthritic models. This review aims to epitomize the role of this high-throughput and multiplexed gene editing method as an analogous therapeutic strategy that could greatly facilitate the clinical development of OA-related treatments since it's reportedly an easy, minimally invasive technique, and a comparatively less painful method for osteoarthritic patients.
Collapse
Affiliation(s)
- Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, China.
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
4
|
Walker AJ, Graham C, Greenwood M, Woodall M, Maeshima R, O’Hara-Wright M, Sanz DJ, Guerrini I, Aldossary AM, O’Callaghan C, Baines DL, Harrison PT, Hart SL. Molecular and functional correction of a deep intronic splicing mutation in CFTR by CRISPR-Cas9 gene editing. Mol Ther Methods Clin Dev 2023; 31:101140. [PMID: 38027060 PMCID: PMC10661860 DOI: 10.1016/j.omtm.2023.101140] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/17/2023] [Indexed: 12/01/2023]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the CFTR gene. The 10th most common mutation, c.3178-2477C>T (3849+10kb C>T), involves a cryptic, intronic splice site. This mutation was corrected in CF primary cells homozygous for this mutation by delivering pairs of guide RNAs (gRNAs) with Cas9 protein in ribonucleoprotein (RNP) complexes that introduce double-strand breaks to flanking sites to excise the 3849+10kb C>T mutation, followed by DNA repair by the non-homologous end-joining pathway, which functions in all cells of the airway epithelium. RNP complexes were delivered to CF basal epithelial cell by a non-viral, receptor-targeted nanocomplex comprising a formulation of targeting peptides and lipids. Canonical CFTR mRNA splicing was, thus, restored leading to the restoration of CFTR protein expression with concomitant restoration of electrophysiological function in airway epithelial air-liquid interface cultures. Off-target editing was not detected by Sanger sequencing of in silico-selected genomic sites with the highest sequence similarities to the gRNAs, although more sensitive unbiased whole genome sequencing methods would be required for possible translational developments. This approach could potentially be used to correct aberrant splicing signals in several other CF mutations and other genetic disorders where deep-intronic mutations are pathogenic.
Collapse
Affiliation(s)
- Amy J. Walker
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Carina Graham
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Miriam Greenwood
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Maximillian Woodall
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
| | - Ruhina Maeshima
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Michelle O’Hara-Wright
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - David J. Sanz
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Ileana Guerrini
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Ahmad M. Aldossary
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Christopher O’Callaghan
- Infection, Immunity & Inflammation Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Deborah L. Baines
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
| | - Patrick T. Harrison
- Department of Physiology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Stephen L. Hart
- Genetics and Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
5
|
Jarallah SJ, Aldossary AM, Tawfik EA, Altamimi RM, Alsharif WK, Alzahrani NM, As Sobeai HM, Qamar W, Alfahad AJ, Alshabibi MA, Alqahtani SH, Alshehri AA, Almughem FA. GL67 lipid-based liposomal formulation for efficient siRNA delivery into human lung cancer cells. Saudi Pharm J 2023; 31:1139-1148. [PMID: 37273265 PMCID: PMC10236467 DOI: 10.1016/j.jsps.2023.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/14/2023] [Indexed: 06/06/2023] Open
Abstract
The efficient delivery of small interfering RNA (siRNA) to the targeted cells significantly affects the regulation of the overexpressed proteins involved in the progression of several genetic diseases. SiRNA molecules in naked form suffer from low internalization across the cell membrane, high susceptibility to degradation by nuclease enzyme and low stability, which hinder their efficacy. Therefore, there is an urge to develop a delivery system that can protect siRNA from degradation and facilitate their uptake across the cell membrane. In this study, the cationic lipid (GL67) was exploited, in addition to DC-Chol and DOPE lipids, to design an efficient liposomal nanocarrier for siRNA delivery. The physiochemical characterizations demonstrated that the molar ratio of 3:1 has proper particle size measurements from 144 nm to 332 nm and zeta potential of -9 mV to 47 mV that depends on the ratio of the GL67 in the liposomal formulation. Gel retardation assay exhibited that increasing the percentage of GL67 in the formulations has a good impact on the encapsulation efficiency compared to DC-Chol. The optimal formulations of the 3:1 M ratio also showed high metabolic activity against A549 cells following a 24 h cell exposure. Flow cytometry findings showed that the highest GL67 lipid ratio (100 % GL67 and 0 % DC-Chol) had the highest percentage of cellular uptake. The lipoplex nanocarriers based on GL67 lipid could potentially influence treating genetic diseases owing to the high internalization efficiency and safety profile.
Collapse
Affiliation(s)
- Somayah J. Jarallah
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Ahmad M. Aldossary
- Wellness and Preventative Medicine Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Essam A. Tawfik
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Reem M. Altamimi
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Wijdan K. Alsharif
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Nouf M. Alzahrani
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Homood M. As Sobeai
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wajhul Qamar
- Pharmacology and Toxicology Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed J. Alfahad
- Bioengineering Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Manal A. Alshabibi
- Healthy Aging Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Sarah H. Alqahtani
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Abdullah A. Alshehri
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Fahad A. Almughem
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| |
Collapse
|
6
|
Amistadi S, Maule G, Ciciani M, Ensinck MM, De Keersmaecker L, Ramalho AS, Guidone D, Buccirossi M, Galietta LJV, Carlon MS, Cereseto A. Functional restoration of a CFTR splicing mutation through RNA delivery of CRISPR adenine base editor. Mol Ther 2023; 31:1647-1660. [PMID: 36895161 PMCID: PMC10277887 DOI: 10.1016/j.ymthe.2023.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/07/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The 2789+5G>A CFTR mutation is a quite frequent defect causing an aberrant splicing and a non-functional CFTR protein. Here we used a CRISPR adenine base editing (ABE) approach to correct the mutation in the absence of DNA double-strand breaks (DSB). To select the strategy, we developed a minigene cellular model reproducing the 2789+5G>A splicing defect. We obtained up to 70% editing in the minigene model by adapting the ABE to the PAM sequence optimal for targeting 2789+5G>A with a SpCas9-NG (NG-ABE). Nonetheless, the on-target base correction was accompanied by secondary (bystander) A-to-G conversions in nearby nucleotides, which affected the wild-type CFTR splicing. To decrease the bystander edits, we used a specific ABE (NG-ABEmax), which was delivered as mRNA. The NG-ABEmax RNA approach was validated in patient-derived rectal organoids and bronchial epithelial cells showing sufficient gene correction to recover the CFTR function. Finally, in-depth sequencing revealed high editing precision genome-wide and allele-specific correction. Here we report the development of a base editing strategy to precisely repair the 2789+5G>A mutation resulting in restoration of the CFTR function, while reducing bystander and off-target activities.
Collapse
Affiliation(s)
- Simone Amistadi
- University of Trento, Department of Computational, Cellular and Integrative Biology, Laboratory of Molecular Virology, 38123 Trento, Italy
| | - Giulia Maule
- University of Trento, Department of Computational, Cellular and Integrative Biology, Laboratory of Molecular Virology, 38123 Trento, Italy.
| | - Matteo Ciciani
- University of Trento, Department of Computational, Cellular and Integrative Biology, Laboratory of Molecular Virology, 38123 Trento, Italy
| | - Marjolein M Ensinck
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Laboratory for Molecular Virology and Gene Therapy, 3000 Leuven, Belgium
| | - Liesbeth De Keersmaecker
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Laboratory for Molecular Virology and Gene Therapy, 3000 Leuven, Belgium
| | - Anabela S Ramalho
- CF Research Lab, Woman and Child Unit, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy
| | | | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy; Department of Translational Medical Sciences, University of Napoli "Federico II," 80138 Napoli, Italy
| | - Marianne S Carlon
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Laboratory for Molecular Virology and Gene Therapy, 3000 Leuven, Belgium; KU Leuven, Department of Chronic Diseases and Metabolism, BREATHE Laboratory, 3000 Leuven, Belgium
| | - Anna Cereseto
- University of Trento, Department of Computational, Cellular and Integrative Biology, Laboratory of Molecular Virology, 38123 Trento, Italy.
| |
Collapse
|
7
|
Li X, Shen L, Deng Z, Huang Z. New treatment for osteoarthr: pbad014itis: Gene therapy. PRECISION CLINICAL MEDICINE 2023; 6:pbad014. [PMID: 37333626 PMCID: PMC10273835 DOI: 10.1093/pcmedi/pbad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Osteoarthritis is a complex degenerative disease that affects the entire joint tissue. Currently, non-surgical treatments for osteoarthritis focus on relieving pain. While end-stage osteoarthritis can be treated with arthroplasty, the health and financial costs associated with surgery have forced the search for alternative non-surgical treatments to delay the progression of osteoarthritis and promote cartilage repair. Unlike traditional treatment, the gene therapy approach allows for long-lasting expression of therapeutic proteins at specific sites. In this review, we summarize the history of gene therapy in osteoarthritis, outlining the common expression vectors (non-viral, viral), the genes delivered (transcription factors, growth factors, inflammation-associated cytokines, non-coding RNAs) and the mode of gene delivery (direct delivery, indirect delivery). We highlight the application and development prospects of the gene editing technology CRISPR/Cas9 in osteoarthritis. Finally, we identify the current problems and possible solutions in the clinical translation of gene therapy for osteoarthritis.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Leyao Shen
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
8
|
Lomunova MA, Gershovich PM. Gene Therapy for Cystic Fibrosis: Recent Advances and Future Prospects. Acta Naturae 2023; 15:20-31. [PMID: 37538805 PMCID: PMC10395777 DOI: 10.32607/actanaturae.11708] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/22/2023] [Indexed: 08/05/2023] Open
Abstract
Gene replacement therapies are novel therapeutic approaches that seek to tackle hereditary diseases caused by a congenital deficiency in a particular gene, when a functional copy of a gene can be delivered to the cells and tissues using various delivery systems. To do this, viral particles carrying a functional copy of the gene of interest and various nonviral gene delivery systems, including liposomes, nanoparticles, etc., can be used. In this review, we discuss the state of current knowledge regarding the molecular mechanisms and types of genetic mutations that lead to cystic fibrosis and highlight recent developments in gene therapy that can be leveraged to correct these mutations and to restore the physiological function of the carrier protein transporting sodium and chlorine ions in the airway epithelial cells. Restoration of carrier protein expression could lead to the normalization of ion and water transport across the membrane and induce a decrease in the viscosity of airway surface fluid, which is one of the pathological manifestations of this disease. This review also summarizes recently published preclinical and clinical data for various gene therapies to allow one to make some conclusions about future prospects for gene therapy in cystic fibrosis treatment.
Collapse
|
9
|
Sufian MA, Ilies MA. Lipid-based nucleic acid therapeutics with in vivo efficacy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1856. [PMID: 36180107 PMCID: PMC10023279 DOI: 10.1002/wnan.1856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/22/2022] [Accepted: 08/30/2022] [Indexed: 03/09/2023]
Abstract
Synthetic vectors for therapeutic nucleic acid delivery are currently competing significantly with their viral counter parts due to their reduced immunogenicity, large payload capacity, and ease of manufacture under GMP-compliant norms. The approval of Onpattro, a lipid-based siRNA therapeutic, and the proven clinical success of two lipid-based COVID-19 vaccines from Pfizer-BioNTech, and Moderna heralded the specific advantages of lipid-based systems among all other synthetic nucleic acid carriers. Lipid-based systems with diverse payloads-plasmid DNA (pDNA), antisense oligonucleotide (ASO), small interfering RNA (siRNA), microRNA (miRNA), small activating RNA (saRNA), and messenger RNA (mRNA)-are now becoming a mature technology, with growing impact in the clinic. Research over four decades identified the key factors determining the therapeutic success of these multi-component systems. Here, we discuss the main nucleic acid-based technologies, presenting their mechanism of action, delivery barriers facing them, the structural properties of the payload as well as the component lipids that regulate physicochemical properties, pharmacokinetics and biodistribution, efficacy, and toxicity of the resultant nanoparticles. We further detail on the formulation parameters, evolution of the manufacturing techniques that generate reproducible and scalable outputs, and key manufacturing aspects that enable control over physicochemical properties of the resultant particles. Preclinical applications of some of these formulations that were successfully translated from in vitro studies to animal models are subsequently discussed. Finally, clinical success and failure of these systems starting from 1993 to present are highlighted, in a holistic literature review focused on lipid-based nucleic acid delivery systems. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials.
Collapse
Affiliation(s)
- Md Abu Sufian
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Marc A. Ilies
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
10
|
Lee JA, Cho A, Huang EN, Xu Y, Quach H, Hu J, Wong AP. Gene therapy for cystic fibrosis: new tools for precision medicine. J Transl Med 2021; 19:452. [PMID: 34717671 PMCID: PMC8556969 DOI: 10.1186/s12967-021-03099-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/01/2021] [Indexed: 12/18/2022] Open
Abstract
The discovery of the Cystic fibrosis (CF) gene in 1989 has paved the way for incredible progress in treating the disease such that the mean survival age of individuals living with CF is now ~58 years in Canada. Recent developments in gene targeting tools and new cell and animal models have re-ignited the search for a permanent genetic cure for all CF. In this review, we highlight some of the more recent gene therapy approaches as well as new models that will provide insight into personalized therapies for CF.
Collapse
Affiliation(s)
- Jin-A Lee
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, 686 Bay Street, PGCRL 16-9420, Toronto, ON, M5G0A4, Canada
| | - Alex Cho
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Elena N Huang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Yiming Xu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Henry Quach
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Jim Hu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
| | - Amy P Wong
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, 686 Bay Street, PGCRL 16-9420, Toronto, ON, M5G0A4, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| |
Collapse
|
11
|
Mercier J, Ruffin M, Corvol H, Guillot L. Gene Therapy: A Possible Alternative to CFTR Modulators? Front Pharmacol 2021; 12:648203. [PMID: 33967785 PMCID: PMC8097140 DOI: 10.3389/fphar.2021.648203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a rare genetic disease that affects several organs, but lung disease is the major cause of morbidity and mortality. The gene responsible for CF, the CFTR (Cystic Fibrosis Transmembrane Conductance Regulator) gene, has been discovered in 1989. Since then, gene therapy i.e., defective gene replacement by a functional one, remained the ultimate goal but unfortunately, it has not yet been achieved. However, patients care and symptomatic treatments considerably increased CF patients’ life expectancy ranging from 5 years old in the 1960s to 40 today. In the last decade, research works on CFTR protein structure and activity led to the development of new drugs which, by readdressing CFTR to the plasma membrane (correctors) or by enhancing its transport activity (potentiators), allow, alone or in combination, an improvement of CF patients’ lung function and quality of life. While expected, it is not yet known whether taking these drugs from an early age and for years will improve the quality of life of CF patients in the long term and further increase their life expectancy. Besides, these molecules are not available (specific variants of CFTR) or accessible (national health policies) for all patients and there is still no curative treatment. Another alternative that could benefit from new technologies, such as gene therapy, is therefore still attractive, although it is not yet offered to patients. Faced with the development of new CFTR correctors and potentiators, the question arises as to whether there is still a place for gene therapy and this is discussed in this perspective.
Collapse
Affiliation(s)
- J Mercier
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| | - M Ruffin
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| | - H Corvol
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France.,Pneumologie Pédiatrique, APHP, Hôpital Trousseau, Paris, France
| | - L Guillot
- Sorbonne Université, Inserm, Centre de Recherche, Saint Antoine, F-75012, Paris, France
| |
Collapse
|
12
|
Sarkar S, Tran N, Soni SK, Nasa Z, Drummond CJ, Conn CE. Cuboplex-Mediated Nonviral Delivery of Functional siRNA to Chinese Hamster Ovary (CHO) Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:2336-2345. [PMID: 33410653 DOI: 10.1021/acsami.0c20956] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Lipid nanoparticles of internal cubic symmetry, termed cuboplexes, are potential nonviral delivery vehicles for gene therapy due to their "topologically active" nature, which may enhance endosomal escape and improve delivery outcomes. In this study, we have used cationic cuboplexes, based on monoolein (MO) doped with a cationic lipid, for the encapsulation and delivery of antisense green fluorescent protein (GFP)-small interfering RNA (siRNA) into Chinese Hamster Ovary (CHO)-GFP cells. Agarose gel electrophoresis has confirmed the successful encapsulation of siRNA within cationic cubosomes, while synchrotron small-angle X-ray scattering (SAXS) demonstrated that the underlying cubic nanostructure of the particles was retained following encapsulation. The cationic cubosomes were shown to be reasonably nontoxic against the CHO-GFP cell line. Fluorescence-activated cell sorting (FACS) provided evidence of the successful transfection to CHO-GFP cells. Knockdown efficiency was strongly linked to the type of cationic lipid used, although all cubosomes had essentially the same internal nanostructure. The gene knockdown efficiency for some cationic cubosomes was shown to be higher than lipofectamine, which is a commercially available liposome-based formulation, while the controlled release of the siRNA from the cubosomes over a 72 h period was observed using confocal microscopy. This combination exemplifies the potential of cationic cuboplexes as a novel, nonviral, controlled-release delivery vector for siRNA.
Collapse
Affiliation(s)
- Sampa Sarkar
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, Victoria 3000, Australia
| | - Nhiem Tran
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, Victoria 3000, Australia
| | - Sarvesh Kumar Soni
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, Victoria 3000, Australia
| | - Zeyad Nasa
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, Victoria 3000, Australia
| | - Calum J Drummond
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, Victoria 3000, Australia
| | - Charlotte E Conn
- School of Science, College of Science, Engineering and Health, RMIT University, 124 La Trobe Street, Melbourne, Victoria 3000, Australia
| |
Collapse
|
13
|
Mukherjee A, MacDonald KD, Kim J, Henderson MI, Eygeris Y, Sahay G. Engineered mutant α-ENaC subunit mRNA delivered by lipid nanoparticles reduces amiloride currents in cystic fibrosis-based cell and mice models. SCIENCE ADVANCES 2020; 6:6/47/eabc5911. [PMID: 33208364 PMCID: PMC7673816 DOI: 10.1126/sciadv.abc5911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/05/2020] [Indexed: 05/02/2023]
Abstract
Cystic fibrosis (CF) results from mutations in the chloride-conducting CF transmembrane conductance regulator (CFTR) gene. Airway dehydration and impaired mucociliary clearance in CF is proposed to result in tonic epithelial sodium channel (ENaC) activity, which drives amiloride-sensitive electrogenic sodium absorption. Decreasing sodium absorption by inhibiting ENaC can reverse airway surface liquid dehydration. Here, we inhibit endogenous heterotrimeric ENaC channels by introducing inactivating mutant ENaC α mRNA (αmutENaC). Lipid nanoparticles carrying αmutENaC were transfected in CF-based airway cells in vitro and in vivo. We observed a significant decrease in macroscopic as well as amiloride-sensitive ENaC currents and an increase in airway surface liquid height in CF airway cells. Similarly, intranasal transfection of αmutENaC mRNA decreased amiloride-sensitive nasal potential difference in CFTRKO mice. These data suggest that mRNA-based ENaC inhibition is a powerful strategy for reducing mucus dehydration and has therapeutic potential for treating CF in all patients, independent of genotype.
Collapse
Affiliation(s)
- Anindit Mukherjee
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR 97201, USA
| | - Kelvin D MacDonald
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR 97201, USA
- Department of Pediatrics, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR 97201, USA
| | - Michael I Henderson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Yulia Eygeris
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR 97201, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR 97201, USA.
- Department of Biomedical Engineering, Robertson Life Sciences Building, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
14
|
Charbe NB, Amnerkar ND, Ramesh B, Tambuwala MM, Bakshi HA, Aljabali AA, Khadse SC, Satheeshkumar R, Satija S, Metha M, Chellappan DK, Shrivastava G, Gupta G, Negi P, Dua K, Zacconi FC. Small interfering RNA for cancer treatment: overcoming hurdles in delivery. Acta Pharm Sin B 2020; 10:2075-2109. [PMID: 33304780 PMCID: PMC7714980 DOI: 10.1016/j.apsb.2020.10.005] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/24/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
In many ways, cancer cells are different from healthy cells. A lot of tactical nano-based drug delivery systems are based on the difference between cancer and healthy cells. Currently, nanotechnology-based delivery systems are the most promising tool to deliver DNA-based products to cancer cells. This review aims to highlight the latest development in the lipids and polymeric nanocarrier for siRNA delivery to the cancer cells. It also provides the necessary information about siRNA development and its mechanism of action. Overall, this review gives us a clear picture of lipid and polymer-based drug delivery systems, which in the future could form the base to translate the basic siRNA biology into siRNA-based cancer therapies.
Collapse
Key Words
- 1,3-propanediol, PEG-b-PDMAEMA-b-Ppy
- 2-propylacrylicacid, PAH-b-PDMAPMA-b-PAH
- APOB, apolipoprotein B
- AQP-5, aquaporin-5
- AZEMA, azidoethyl methacrylate
- Atufect01, β-l-arginyl-2,3-l-diaminopropionicacid-N-palmityl-N-oleyl-amide trihydrochloride
- AuNPs, gold nanoparticles
- B-PEI, branched polyethlenimine
- BMA, butyl methacrylate
- CFTR, cystic fibrosis transmembrane conductance regulator gene
- CHEMS, cholesteryl hemisuccinate
- CHOL, cholesterol
- CMC, critical micelles concentration
- Cancer
- DC-Chol, 3β-[N-(N′,N′-dimethylaminoethane)carbamoyl]cholesterol
- DMAEMA, 2-dimethylaminoethyl methacrylate
- DNA, deoxyribonucleic acid
- DOPC, dioleylphosphatidyl choline
- DOPE, dioleylphosphatidyl ethanolamine
- DOTAP, N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methyl-sulfate
- DOTMA, N-[1-(2,3-dioleyloxy)propy]-N,N,N-trimethylammoniumchloride
- DOX, doxorubicin
- DSGLA, N,N-dis-tearyl-N-methyl-N-2[N′-(N2-guanidino-l-lysinyl)] aminoethylammonium chloride
- DSPC, 1,2-distearoyl-sn-glycero-3-phosphocholine
- DSPE, 1,2-distearoyl-sn-glycero-3-phosphorylethanolamine
- DSPE-MPEG, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (ammonium salt)
- DSPE-PEG-Mal: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[maleimide(polyethylene glycol)-2000] (mmmonium salt), EPR
- Liposomes
- Micelles
- N-acetylgalactosamine, HIF-1α
- Nanomedicine
- PE-PCL-b-PNVCL, pentaerythritol polycaprolactone-block-poly(N-vinylcaprolactam)
- PLA, poly-l-arginine
- PLGA, poly lactic-co-glycolic acid
- PLK-1, polo-like kinase 1
- PLL, poly-l-lysine
- PPES-b-PEO-b-PPES, poly(4-(phenylethynyl)styrene)-block-PEO-block-poly(4-(phenylethynyl)styrene)
- PTX, paclitaxel
- PiRNA, piwi-interacting RNA
- Polymer
- RES, reticuloendothelial system
- RGD, Arg-Gly-Asp peptide
- RISC, RNA-induced silencing complex
- RNA, ribonucleic acid
- RNAi, RNA interference
- RNAse III, ribonuclease III enzyme
- SEM, scanning electron microscope
- SNALP, stable nucleic acid-lipid particles
- SiRNA, short interfering rNA
- Small interfering RNA (siRNA)
- S–Au, thio‒gold
- TCC, transitional cell carcinoma
- TEM, transmission electron microscopy
- Tf, transferrin
- Trka, tropomyosin receptor kinase A
- USPIO, ultra-small superparamagnetic iron oxide nanoparticles
- UV, ultraviolet
- VEGF, vascular endothelial growth factor
- ZEBOV, Zaire ebola virus
- enhanced permeability and retention, Galnac
- hypoxia-inducible factor-1α, KSP
- kinesin spindle protein, LDI
- lipid-protamine-DNA/hyaluronic acid, MDR
- lysine ethyl ester diisocyanate, LPD/LPH
- messenger RNA, MTX
- methotrexate, NIR
- methoxy polyethylene glycol-polycaprolactone, mRNA
- methoxypoly(ethylene glycol), MPEG-PCL
- micro RNA, MPEG
- multiple drug resistance, MiRNA
- nanoparticle, NRP-1
- near-infrared, NP
- neuropilin-1, PAA
- poly(N,N-dimethylacrylamide), PDO
- poly(N-isopropyl acrylamide), pentaerythritol polycaprolactone-block-poly(N-isopropylacrylamide)
- poly(acrylhydrazine)-block-poly(3-dimethylaminopropyl methacrylamide)-block-poly(acrylhydrazine), PCL
- poly(ethylene glycol)-block-poly(2-dimethylaminoethyl methacrylate)-block poly(pyrenylmethyl methacrylate), PEG-b-PLL
- poly(ethylene glycol)-block-poly(l-lysine), PEI
- poly(ethylene oxide)-block-poly(2-(diethylamino)ethyl methacrylate)-stat-poly(methoxyethyl methacrylate), PEO-b-PCL
- poly(ethylene oxide)-block-poly(Ε-caprolactone), PE-PCL-b-PNIPAM
- poly(Ε-caprolactone), PCL-PEG
- poly(Ε-caprolactone)-polyethyleneglycol-poly(l-histidine), PCL-PEI
- polycaprolactone-polyethyleneglycol, PCL-PEG-PHIS
- polycaprolactone-polyethylenimine, PDMA
- polyethylenimine, PEO-b-P(DEA-Stat-MEMA
Collapse
Affiliation(s)
- Nitin Bharat Charbe
- Departamento de Quimica Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Sri Adichunchunagiri College of Pharmacy, Sri Adichunchunagiri University, BG Nagar, Karnataka 571418, India
| | - Nikhil D. Amnerkar
- Adv V. R. Manohar Institute of Diploma in Pharmacy, Nagpur, Maharashtra 441110, India
| | - B. Ramesh
- Sri Adichunchunagiri College of Pharmacy, Sri Adichunchunagiri University, BG Nagar, Karnataka 571418, India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Hamid A. Bakshi
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Alaa A.A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan
| | - Saurabh C. Khadse
- Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Dist. Dhule, Maharashtra 425 405, India
| | - Rajendran Satheeshkumar
- Departamento de Quimica Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Saurabh Satija
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411 Punjab, India
| | - Meenu Metha
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411 Punjab, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Garima Shrivastava
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, New Delhi 110016, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur 302017, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW 2308, Australia
| | - Flavia C. Zacconi
- Departamento de Quimica Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 4860, Chile
| |
Collapse
|
15
|
Somayaji R, Nichols DP, Bell SC. Cystic fibrosis - Ten promising therapeutic approaches in the current era of care. Expert Opin Investig Drugs 2020; 29:1107-1124. [PMID: 32744089 DOI: 10.1080/13543784.2020.1805733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic disease affecting multiple organ systems. Research and innovations in novel therapeutic agents and health care delivery have resulted in dramatic improvements in quality of life and survival for people with CF. Despite this, significant disease burden persists for many and this is compounded by disparities in treatment access and care which globally necessitates further work to improve outcomes. Because of the advent of numerous therapies which include gene-targeted modulators in parallel with specialized care delivery models, innovative efforts continue. AREAS COVERED In this review, we discuss the available data on investigational agents in clinical development and currently available treatments for CF. We also evaluate approaches to care delivery, consider treatment gaps, and propose future directions for advancement. EXPERT OPINION Since the discovery of the CF gene, CFTR modulators have provided a hallmark of success, even though it was thought not previously possible. This has led to reinvigorated efforts and innovations in treatment approaches and care delivery. Numerous challenges remain because of genetic and phenotypic heterogeneity, access issues, and therapeutic costs, but the collaborative approach between stakeholders for continued innovation fuels optimism. Abbreviations: CF cystic fibrosis; CFF Cystic Fibrosis Foundation (USA); CFTR cystic fibrosis transmembrane regulator; CRISPR clustered regularly interspaced short palindromic repeats; COX cyclo oxygenase; FDA US Food and Drug Administration; FEV1% forced expiratory volume in one second % predicted; F508del deletion of phenylalanine (F) in the 508th position (most common mutation); G551D substitution of the amino acid glycine by aspartate at position 551 in the nucleotide binding domain-1 of the CFTR gene; LMIC low- and middle-income country; LTB4 leukotriene B4; MDT multi-disciplinary care team; NO nitric oxide; NSAIDs non-steroidal anti-inflammatory drugs; SLPI secretory leukocyte protease inhibitor.
Collapse
Affiliation(s)
- Ranjani Somayaji
- Departments of Medicine; Microbiology, Immunology & Infectious Disease; Community Health Sciences, University of Calgary , Calgary, AB, Canada.,Snyder Institute for Chronic Diseases , Calgary, AB, Canada.,O'Brien Institute for Public Health , Calgary, AB, Canada
| | - Dave P Nichols
- Department of Pediatrics, Seattle Children's Hospital , Seattle, WA, USA.,Department of Pediatrics, University of Washington , Seattle, WA, USA.,Seattle Children's Research Institute , Seattle, WA, USA
| | - Scott C Bell
- Department of Thoracic Medicine, The Prince Charles Hospital , Brisbane, QLD, Australia.,Children's Health Research Centre, Faculty of Medicine, The University of Queensland , Brisbane, QLD, Australia.,Translational Research Institute , Brisbane, QLD, Australia
| |
Collapse
|
16
|
Maule G, Arosio D, Cereseto A. Gene Therapy for Cystic Fibrosis: Progress and Challenges of Genome Editing. Int J Mol Sci 2020; 21:E3903. [PMID: 32486152 PMCID: PMC7313467 DOI: 10.3390/ijms21113903] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 02/07/2023] Open
Abstract
Since the early days of its conceptualization and application, human gene transfer held the promise of a permanent solution to genetic diseases including cystic fibrosis (CF). This field went through alternated periods of enthusiasm and distrust. The development of refined technologies allowing site specific modification with programmable nucleases highly revived the gene therapy field. CRISPR nucleases and derived technologies tremendously facilitate genome manipulation offering diversified strategies to reverse mutations. Here we discuss the advancement of gene therapy, from therapeutic nucleic acids to genome editing techniques, designed to reverse genetic defects in CF. We provide a roadmap through technologies and strategies tailored to correct different types of mutations in the cystic fibrosis transmembrane regulator (CFTR) gene, and their applications for the development of experimental models valuable for the advancement of CF therapies.
Collapse
Affiliation(s)
- Giulia Maule
- Department of Cellular Computational Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy;
- National Council of Research, CNR, 38123 Trento, Italy;
| | | | - Anna Cereseto
- Department of Cellular Computational Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy;
| |
Collapse
|
17
|
Keil TWM, Baldassi D, Merkel OM. T-cell targeted pulmonary siRNA delivery for the treatment of asthma. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1634. [PMID: 32267622 DOI: 10.1002/wnan.1634] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 12/31/2022]
Abstract
Despite the large number of drugs available for the treatment of asthma, in 5-10% of the patients this disease is not well controlled. While most treatments palliate symptoms, those suffering from severe and uncontrolled asthma could benefit more from a therapeutic approach addressing the root problem. An siRNA-based therapy targeting the transcription factor GATA3 in activated T helper cells subtype 2 (TH 2 cells), one of the key upstream factors involved in asthma, could therefore represent a promising strategy. However, the difficult-to-transfect cell type has not extensively been explored for nucleic acid therapeutics. In this regard, our group first identified a suitable pathway, that is, transferrin receptor mediated uptake, to target efficiently and specifically activated TH 2 cells with a transferrin-polyethyleneimine (PEI) conjugate which forms polyplexes with siRNA. This system, despite efficient uptake in activated T cells (ATCs) in vivo, suffered from poor endosomal release and was later improved by a combination with a melittin-PEI conjugate. The new formulation showed improved endosomal escape and gene silencing efficacy. Additionally, in order to develop a clinically relevant dosage form for pulmonary delivery of siRNA we have lately focused on a dry powder formulation by spray drying (SD) for the production of inhalable nano-in-microparticles. In proof-of-concept experiments, DNA/PEI polyplexes were used in order to implement analytics and engineer process parameters to pave the way for SD also siRNA containing polyplexes and more sophisticated systems in general. Ultimately, our efforts are devoted to the development of a novel treatment of asthma that can be translated from bench to bedside and are reviewed and discussed here in the context of the current literature. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Respiratory Disease Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Tobias W M Keil
- Pharmaceutical Technology and Biopharmaceutics, LMU Munich, Munich, Germany
| | - Domizia Baldassi
- Pharmaceutical Technology and Biopharmaceutics, LMU Munich, Munich, Germany
| | - Olivia M Merkel
- Pharmaceutical Technology and Biopharmaceutics, LMU Munich, Munich, Germany
| |
Collapse
|
18
|
Ehexige E, Ganbold T, Yu X, Han S, Baigude H. Design of Peptidomimetic Functionalized Cholesterol Based Lipid Nanoparticles for Efficient Delivery of Therapeutic Nucleic Acids. Molecules 2019; 24:E3413. [PMID: 31546908 PMCID: PMC6767268 DOI: 10.3390/molecules24183413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 11/29/2022] Open
Abstract
Lipid nanoparticles (LNP) are the most potent carriers for the delivery of nucleic acid-based therapeutics. The first FDA approved a short interfering RNA (siRNA) drug that uses a cationic LNP system for the delivery of siRNA against human transthyretin (hTTR). However, preparation of such LNP involves tedious multi-step synthesis with relatively low yields. In the present study, we synthesized cationic peptidomimetic functionalized cholesterol (denote Chorn) in straightforward chemical approaches with high yield. When formulated with helper lipids, Chorn LNPs complexed with siRNA to form nanoparticles with an average diameter of 150 nm to 200 nm. Chorn LNP mediated transfection of a green fluorescence protein (GFP) expressing plasmid resulted in 60% GFP positive cells. Moreover, Chorn LNP delivered siRNA against polo-like kinase 1 (Plk1), a disease related gene in cancer cells and efficiently suppressed the expression of the gene, resulting in significant morphological changes in the cell nuclei. Our data suggested that cholesterol based cationic LNP, prepared through a robust chemical strategy, may provide a promising siRNA delivery system.
Collapse
Affiliation(s)
- Ehexige Ehexige
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, China.
| | - Tsogzolmaa Ganbold
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, China.
| | - Xiang Yu
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, China.
| | - Shuqin Han
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, China.
| | - Huricha Baigude
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, China.
| |
Collapse
|
19
|
Buck J, Grossen P, Cullis PR, Huwyler J, Witzigmann D. Lipid-Based DNA Therapeutics: Hallmarks of Non-Viral Gene Delivery. ACS NANO 2019; 13:3754-3782. [PMID: 30908008 DOI: 10.1021/acsnano.8b07858] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Gene therapy is a promising strategy for the treatment of monogenic disorders. Non-viral gene delivery systems including lipid-based DNA therapeutics offer the opportunity to deliver an encoding gene sequence specifically to the target tissue and thus enable the expression of therapeutic proteins in diseased cells. Currently, available gene delivery approaches based on DNA are inefficient and require improvements to achieve clinical utility. In this Review, we discuss state-of-the-art lipid-based DNA delivery systems that have been investigated in a preclinical setting. We emphasize factors influencing the delivery and subsequent gene expression in vitro, ex vivo, and in vivo. In addition, we cover aspects of nanoparticle engineering and optimization for DNA therapeutics. Finally, we highlight achievements of lipid-based DNA therapies in clinical trials.
Collapse
Affiliation(s)
- Jonas Buck
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
- Department of Biochemistry and Molecular Biology , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| |
Collapse
|
20
|
Pranke I, Golec A, Hinzpeter A, Edelman A, Sermet-Gaudelus I. Emerging Therapeutic Approaches for Cystic Fibrosis. From Gene Editing to Personalized Medicine. Front Pharmacol 2019; 10:121. [PMID: 30873022 PMCID: PMC6400831 DOI: 10.3389/fphar.2019.00121] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/31/2019] [Indexed: 12/13/2022] Open
Abstract
An improved understanding of the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) protein structure and the consequences of CFTR gene mutations have allowed the development of novel therapies targeting specific defects underlying CF. Some strategies are mutation specific and have already reached clinical development; some strategies include a read-through of the specific premature termination codons (read-through therapies, nonsense mediated decay pathway inhibitors for Class I mutations); correction of CFTR folding and trafficking to the apical plasma membrane (correctors for Class II mutations); and an increase in the function of CFTR channel (potentiators therapy for Class III mutations and any mutant with a residual function located at the membrane). Other therapies that are in preclinical development are not mutation specific and include gene therapy to edit the genome and stem cell therapy to repair the airway tissue. These strategies that are directed at the basic CF defects are now revolutionizing the treatment for patients and should positively impact their survival rates.
Collapse
Affiliation(s)
- Iwona Pranke
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | - Anita Golec
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | - Alexandre Hinzpeter
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | - Aleksander Edelman
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France
| | - Isabelle Sermet-Gaudelus
- INSERM U 1151, Institut Necker Enfants Malades, Université Paris Descartes, Paris, France.,Centre de Référence Maladie Rare, Mucoviscidose et Maladies de CFTR, Paris, France.,Faculté de Médecine, Université Paris Descartes, Paris, France
| |
Collapse
|
21
|
Abstract
Calpain is an intracellular Ca2+-dependent non-lysosomal cysteine protease expressed ubiquitously in mammals. In endothelial cells, dysregulation of calpain has been shown to be involved in a wide variety of pathological conditions such as angiogenesis, vascular inflammation, and diabetes. Cell- or tissue-targeted in vivo delivery of small interfering RNA (siRNA) is a powerful research tool in the analysis of protein function and has been proposed as an attractive therapeutic modality that is applicable against a large number of human diseases including cancer. In this chapter we describe a method to knockdown calpain 1 in mouse pulmonary vascular endothelium using delivery of siRNA/cationic liposome complex. This technique results in a greater than 80% reduction in calpain 1 protein levels 48 h after a single i.v. injection of calpain 1 siRNA (0.5 mg siRNA/kg)/cationic liposome complex. We also describe confocal imaging to verify the loss of calpain 1 expression in pulmonary microvessel endothelial cells and application of this technique in the mouse model of ventilator-induced lung injury.
Collapse
|
22
|
Zhang XP, Zhang WT, Qiu Y, Ju MJ, Tu GW, Luo Z. Understanding Gene Therapy in Acute Respiratory Distress Syndrome. Curr Gene Ther 2019; 19:93-99. [PMID: 31267871 DOI: 10.2174/1566523219666190702154817] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/07/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
Acute Respiratory Distress Syndrome (ARDS) and its complications remain lifethreatening conditions for critically ill patients. The present therapeutic strategies such as prone positioning ventilation strategies, nitric oxide inhalation, restrictive intravenous fluid management, and extracorporeal membrane oxygenation (ECMO) do not contribute much to improving the mortality of ARDS. The advanced understanding of the pathophysiology of acute respiratory distress syndrome suggests that gene-based therapy may be an innovative method for this disease. Many scientists have made beneficial attempts to regulate the immune response genes of ARDS, maintain the normal functions of alveolar epithelial cells and endothelial cells, and inhibit the fibrosis and proliferation of ARDS. Limitations to effective pulmonary gene therapy still exist, including the security of viral vectors and the pulmonary defense mechanisms against inhaled particles. Here, we summarize and review the mechanism of gene therapy for acute respiratory distress syndrome and its application.
Collapse
Affiliation(s)
- Xue-Peng Zhang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Wei-Tao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, No. 179 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Yue Qiu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Min-Jie Ju
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Guo-Wei Tu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Zhe Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
- Department of Critical Care Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, No. 668 Jinghu Road, Huli District, Xiamen 361015, China
| |
Collapse
|
23
|
Cooney AL, McCray PB, Sinn PL. Cystic Fibrosis Gene Therapy: Looking Back, Looking Forward. Genes (Basel) 2018; 9:genes9110538. [PMID: 30405068 PMCID: PMC6266271 DOI: 10.3390/genes9110538] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 01/02/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that encodes a cAMP-regulated anion channel. Although CF is a multi-organ system disease, most people with CF die of progressive lung disease that begins early in childhood and is characterized by chronic bacterial infection and inflammation. Nearly 90% of people with CF have at least one copy of the ΔF508 mutation, but there are hundreds of CFTR mutations that result in a range of disease severities. A CFTR gene replacement approach would be efficacious regardless of the disease-causing mutation. After the discovery of the CFTR gene in 1989, the in vitro proof-of-concept for gene therapy for CF was quickly established in 1990. In 1993, the first of many gene therapy clinical trials attempted to rescue the CF defect in airway epithelia. Despite the initial enthusiasm, there is still no FDA-approved gene therapy for CF. Here we discuss the history of CF gene therapy, from the discovery of the CFTR gene to current state-of-the-art gene delivery vector designs. While implementation of CF gene therapy has proven more challenging than initially envisioned; thanks to continued innovation, it may yet become a reality.
Collapse
Affiliation(s)
- Ashley L Cooney
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Paul B McCray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| | - Patrick L Sinn
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
24
|
Kim SB, Lee K, Raj MS, Lee B, Reeder JT, Koo J, Hourlier-Fargette A, Bandodkar AJ, Won SM, Sekine Y, Choi J, Zhang Y, Yoon J, Kim BH, Yun Y, Lee S, Shin J, Kim J, Ghaffari R, Rogers JA. Soft, Skin-Interfaced Microfluidic Systems with Wireless, Battery-Free Electronics for Digital, Real-Time Tracking of Sweat Loss and Electrolyte Composition. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802876. [PMID: 30300469 DOI: 10.1002/smll.201802876] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/12/2018] [Indexed: 05/14/2023]
Abstract
Sweat excretion is a dynamic physiological process that varies with body position, activity level, environmental factors, and health status. Conventional means for measuring the properties of sweat yield accurate results but their requirements for sampling and analytics do not allow for use in the field. Emerging wearable devices offer significant advantages over existing approaches, but each has significant drawbacks associated with bulk and weight, inability to quantify volumetric sweat rate and loss, robustness, and/or inadequate accuracy in biochemical analysis. This paper presents a thin, miniaturized, skin-interfaced microfluidic technology that includes a reusable, battery-free electronics module for measuring sweat conductivity and rate in real-time using wireless power from and data communication to electronic devices with capabilities in near field communications (NFC), including most smartphones. The platform exploits ultrathin electrodes integrated within a collection of microchannels as interfaces to circuits that leverage NFC protocols. The resulting capabilities are complementary to those of previously reported colorimetric strategies. Systematic studies of these combined microfluidic/electronic systems, accurate correlations of measurements performed with them to those of laboratory standard instrumentation, and field tests on human subjects exercising and at rest establish the key operational features and their utility in sweat analytics.
Collapse
Affiliation(s)
- Sung Bong Kim
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - KunHyuck Lee
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Milan S Raj
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
| | - Boram Lee
- Department of Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Jonathan T Reeder
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Jahyun Koo
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Aurélie Hourlier-Fargette
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Amay J Bandodkar
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Sang Min Won
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yurina Sekine
- Materials Sciences Research Center, Japan Atomic Energy Agency, Tokai, Ibaraki, 319-1195, Japan
| | - Jungil Choi
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yi Zhang
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Jangryeol Yoon
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Bong Hoon Kim
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
| | - Yeojeong Yun
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Seojin Lee
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Biomedical, Biological, and Chemical Engineering, University of Missouri, Columbia, MO, 65211, USA
| | - Jiho Shin
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jeonghyun Kim
- Department of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Roozbeh Ghaffari
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Electronics Convergence Engineering, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - John A Rogers
- Center for Bio-Integrated Electronics at the Simpson, Querrey Institute for BioNanotechnology, Northwestern University, Evanston, IL, 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Electronics Convergence Engineering, Kwangwoon University, Seoul, 01897, Republic of Korea
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Chemistry, Northwestern University, Evanston, IL, 60208, USA
- Department of Electrical Engineering and Computer Science, Northwestern University, Evanston, IL, 60208, USA
- Department of Neurological Surgery, Northwestern University, Evanston, IL, 60208, USA
- Simpson Querrey Institute for Nano/Biotechnology, McCormick School of Engineering and Feinberg, School of Medicine, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
25
|
McElvaney OJ, Gunaratnam C, McElvaney OF, Bagwe I, Reeves EP, McElvaney NG. Emerging pharmacotherapies in cystic fibrosis. Expert Rev Respir Med 2018; 12:843-855. [DOI: 10.1080/17476348.2018.1512409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Oliver J McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Cedric Gunaratnam
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Oisin Fiachra McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Isha Bagwe
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
26
|
Robinson E, MacDonald KD, Slaughter K, McKinney M, Patel S, Sun C, Sahay G. Lipid Nanoparticle-Delivered Chemically Modified mRNA Restores Chloride Secretion in Cystic Fibrosis. Mol Ther 2018; 26:2034-2046. [PMID: 29910178 PMCID: PMC6094356 DOI: 10.1016/j.ymthe.2018.05.014] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 05/12/2018] [Accepted: 05/12/2018] [Indexed: 12/14/2022] Open
Abstract
The promise of gene therapy for the treatment of cystic fibrosis has yet to be fully clinically realized despite years of effort toward correcting the underlying genetic defect in the cystic fibrosis transmembrane conductance regulator (CFTR). mRNA therapy via nanoparticle delivery represents a powerful technology for the transfer of genetic material to cells with large, widespread populations, such as airway epithelia. We deployed a clinically relevant lipid-based nanoparticle (LNP) for packaging and delivery of large chemically modified CFTR mRNA (cmCFTR) to patient-derived bronchial epithelial cells, resulting in an increase in membrane-localized CFTR and rescue of its primary function as a chloride channel. Furthermore, nasal application of LNP-cmCFTR restored CFTR-mediated chloride secretion to conductive airway epithelia in CFTR knockout mice for at least 14 days. On day 3 post-transfection, CFTR activity peaked, recovering up to 55% of the net chloride efflux characteristic of healthy mice. This magnitude of response is superior to liposomal CFTR DNA delivery and is comparable with outcomes observed in the currently approved drug ivacaftor. LNP-cmRNA-based systems represent a powerful platform technology for correction of cystic fibrosis and other monogenic disorders.
Collapse
Affiliation(s)
- Ema Robinson
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Kelvin D MacDonald
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA; Department of Pediatrics, School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kai Slaughter
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Madison McKinney
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA
| | - Conroy Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA; Department of Radiation Medicine, School of Medicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR 97201, USA; Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97201, USA.
| |
Collapse
|
27
|
Campani V, Giarra S, De Rosa G. Lipid-based core-shell nanoparticles: Evolution and potentialities in drug delivery. OPENNANO 2018. [DOI: 10.1016/j.onano.2017.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
28
|
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic disorder that results in a multi-organ disease with progressive respiratory decline that ultimately leads to premature death. CF is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which codes for the CFTR anion channel. Established CF treatments target downstream manifestations of the primary genetic defect, including pulmonary and nutritional interventions. Areas covered: CFTR modulators are novel therapies that improve the function of CFTR, and have been approved in the past five years to mitigate the effects of several CF-disease causing mutations. This review summarizes currently approved CFTR modulators and discusses emerging modulator therapies in phase II and III clinical trials described on clinical trials.gov as of April, 2017. Results of relevant trials reported in peer-reviewed journals in Pubmed, scientific conference abstracts and sponsor press releases available as of November, 2017 are included. Expert opinion: The current scope of CF therapeutic development is robust and CFTR modulators have demonstrated significant benefit to patients with specific CFTR mutations. We anticipate that in the future healthcare providers will be faced with a different treatment paradigm, initiating CFTR-directed therapies well before the onset of progressive lung disease.
Collapse
Affiliation(s)
- Kristin M Hudock
- a Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine , University of Cincinnati , Cincinnati , OH , USA.,b Division of Pulmonary Biology, Department of Pediatrics , Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA
| | - John Paul Clancy
- c Division of Pulmonary Medicine, Department of Pediatrics , Cincinnati Children's Hospital Medical Center , Cincinnati , OH , USA
| |
Collapse
|
29
|
da Silva AL, Cruz FF, Rocco PRM, Morales MM. New perspectives in nanotherapeutics for chronic respiratory diseases. Biophys Rev 2017; 9:793-803. [PMID: 28914424 PMCID: PMC5662054 DOI: 10.1007/s12551-017-0319-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
According to the World Health Organization (WHO), hundreds of millions of people of all ages and in all countries suffer from chronic respiratory diseases, with particular negative consequences such as poor health-related quality of life, impaired work productivity, and limitations in the activities of daily living. Chronic obstructive pulmonary disease, asthma, occupational lung diseases (such as silicosis), cystic fibrosis, and pulmonary arterial hypertension are the most common of these diseases, and none of them are curable with current therapies. The advent of nanotechnology holds great therapeutic promise for respiratory conditions, because non-viral vectors are able to overcome the mucus and lung remodeling barriers, increasing pharmacologic and therapeutic potency. It has been demonstrated that the extent of pulmonary nanoparticle uptake depends not only on the physical and chemical features of nanoparticles themselves, but also on the health status of the organism; thus, the huge diversity in nanotechnology could revolutionize medicine, but safety assessment is a challenging task. Within this context, the present review discusses some of the major new perspectives in nanotherapeutics for lung disease and highlights some of the most recent studies in the field.
Collapse
Affiliation(s)
- Adriana Lopes da Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcelo Marcos Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
30
|
Sondhi D, Stiles KM, De BP, Crystal RG. Genetic Modification of the Lung Directed Toward Treatment of Human Disease. Hum Gene Ther 2017; 28:3-84. [PMID: 27927014 DOI: 10.1089/hum.2016.152] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Genetic modification therapy is a promising therapeutic strategy for many diseases of the lung intractable to other treatments. Lung gene therapy has been the subject of numerous preclinical animal experiments and human clinical trials, for targets including genetic diseases such as cystic fibrosis and α1-antitrypsin deficiency, complex disorders such as asthma, allergy, and lung cancer, infections such as respiratory syncytial virus (RSV) and Pseudomonas, as well as pulmonary arterial hypertension, transplant rejection, and lung injury. A variety of viral and non-viral vectors have been employed to overcome the many physical barriers to gene transfer imposed by lung anatomy and natural defenses. Beyond the treatment of lung diseases, the lung has the potential to be used as a metabolic factory for generating proteins for delivery to the circulation for treatment of systemic diseases. Although much has been learned through a myriad of experiments about the development of genetic modification of the lung, more work is still needed to improve the delivery vehicles and to overcome challenges such as entry barriers, persistent expression, specific cell targeting, and circumventing host anti-vector responses.
Collapse
Affiliation(s)
- Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Katie M Stiles
- Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Bishnu P De
- Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| |
Collapse
|
31
|
Inflammasomes in the lung. Mol Immunol 2017; 86:44-55. [PMID: 28129896 DOI: 10.1016/j.molimm.2017.01.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 12/11/2022]
Abstract
Innate immune responses act as first line defences upon exposure to potentially noxious stimuli. The innate immune system has evolved numerous intracellular and extracellular receptors that undertake surveillance for potentially damaging particulates. Inflammasomes are intracellular innate immune multiprotein complexes that form and are activated following interaction with these stimuli. Inflammasome activation leads to the cleavage of pro-IL-1β and release of the pro-inflammatory cytokine, IL-1β, which initiates acute phase pro-inflammatory responses, and other responses are also involved (IL-18, pyroptosis). However, excessive activation of inflammasomes can result in chronic inflammation, which has been implicated in a range of chronic inflammatory diseases. The airways are constantly exposed to a wide variety of stimuli. Inflammasome activation and downstream responses clears these stimuli. However, excessive activation may drive the pathogenesis of chronic respiratory diseases such as severe asthma and chronic obstructive pulmonary disease. Thus, there is currently intense interest in the role of inflammasomes in chronic inflammatory lung diseases and in their potential for therapeutic targeting. Here we review the known associations between inflammasome-mediated responses and the development and exacerbation of chronic lung diseases.
Collapse
|
32
|
Alton EWFW, Beekman JM, Boyd AC, Brand J, Carlon MS, Connolly MM, Chan M, Conlon S, Davidson HE, Davies JC, Davies LA, Dekkers JF, Doherty A, Gea-Sorli S, Gill DR, Griesenbach U, Hasegawa M, Higgins TE, Hironaka T, Hyndman L, McLachlan G, Inoue M, Hyde SC, Innes JA, Maher TM, Moran C, Meng C, Paul-Smith MC, Pringle IA, Pytel KM, Rodriguez-Martinez A, Schmidt AC, Stevenson BJ, Sumner-Jones SG, Toshner R, Tsugumine S, Wasowicz MW, Zhu J. Preparation for a first-in-man lentivirus trial in patients with cystic fibrosis. Thorax 2016; 72:137-147. [PMID: 27852956 PMCID: PMC5284333 DOI: 10.1136/thoraxjnl-2016-208406] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 01/03/2023]
Abstract
We have recently shown that non-viral gene therapy can stabilise the decline of lung function in patients with cystic fibrosis (CF). However, the effect was modest, and more potent gene transfer agents are still required. Fuson protein (F)/Hemagglutinin/Neuraminidase protein (HN)-pseudotyped lentiviral vectors are more efficient for lung gene transfer than non-viral vectors in preclinical models. In preparation for a first-in-man CF trial using the lentiviral vector, we have undertaken key translational preclinical studies. Regulatory-compliant vectors carrying a range of promoter/enhancer elements were assessed in mice and human air–liquid interface (ALI) cultures to select the lead candidate; cystic fibrosis transmembrane conductance receptor (CFTR) expression and function were assessed in CF models using this lead candidate vector. Toxicity was assessed and ‘benchmarked’ against the leading non-viral formulation recently used in a Phase IIb clinical trial. Integration site profiles were mapped and transduction efficiency determined to inform clinical trial dose-ranging. The impact of pre-existing and acquired immunity against the vector and vector stability in several clinically relevant delivery devices was assessed. A hybrid promoter hybrid cytosine guanine dinucleotide (CpG)- free CMV enhancer/elongation factor 1 alpha promoter (hCEF) consisting of the elongation factor 1α promoter and the cytomegalovirus enhancer was most efficacious in both murine lungs and human ALI cultures (both at least 2-log orders above background). The efficacy (at least 14% of airway cells transduced), toxicity and integration site profile supports further progression towards clinical trial and pre-existing and acquired immune responses do not interfere with vector efficacy. The lead rSIV.F/HN candidate expresses functional CFTR and the vector retains 90–100% transduction efficiency in clinically relevant delivery devices. The data support the progression of the F/HN-pseudotyped lentiviral vector into a first-in-man CF trial in 2017.
Collapse
Affiliation(s)
- Eric W F W Alton
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Jeffery M Beekman
- Department of Pediatric Pulmonology, Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | - A Christopher Boyd
- Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - June Brand
- Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK.,Lung Pathology Unit, Department of Airway Disease Infection, NHLI, Imperial College London, London, UK
| | - Marianne S Carlon
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Brussels, Belgium
| | - Mary M Connolly
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Mario Chan
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Sinead Conlon
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Heather E Davidson
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Jane C Davies
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Lee A Davies
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Johanna F Dekkers
- Department of Pediatric Pulmonology, Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | - Ann Doherty
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Sabrina Gea-Sorli
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Deborah R Gill
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Uta Griesenbach
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | | | - Tracy E Higgins
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | | | - Laura Hyndman
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Gerry McLachlan
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Roslin Institute & R(D)SVS, University of Edinburgh, Midlothian, UK
| | - Makoto Inoue
- ID Pharme Co. Ltd. (DNAVEC Center), Tsukuba, Japan
| | - Stephen C Hyde
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - J Alastair Innes
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Toby M Maher
- Fibrosis Research Group, Inflammation, Repair & Development Section, National Heart and Lung Institute, Sir Alexander Fleming Building, Imperial College, London, UK
| | - Caroline Moran
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Cuixiang Meng
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Michael C Paul-Smith
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Ian A Pringle
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Kamila M Pytel
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Andrea Rodriguez-Martinez
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | | | - Barbara J Stevenson
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Stephanie G Sumner-Jones
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Richard Toshner
- Fibrosis Research Group, Inflammation, Repair & Development Section, National Heart and Lung Institute, Sir Alexander Fleming Building, Imperial College, London, UK
| | | | - Marguerite W Wasowicz
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Jie Zhu
- Lung Pathology Unit, Department of Airway Disease Infection, NHLI, Imperial College London, London, UK
| |
Collapse
|
33
|
|
34
|
Alton EWFW, Boyd AC, Davies JC, Gill DR, Griesenbach U, Harrison PT, Henig N, Higgins T, Hyde SC, Innes JA, Korman MSD. Genetic medicines for CF: Hype versus reality. Pediatr Pulmonol 2016; 51:S5-S17. [PMID: 27662105 DOI: 10.1002/ppul.23543] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 12/19/2022]
Abstract
Since identification of the CFTR gene over 25 years ago, gene therapy for cystic fibrosis (CF) has been actively developed. More recently gene therapy has been joined by other forms of "genetic medicines" including mRNA delivery, as well as genome editing and mRNA repair-based strategies. Proof-of-concept that gene therapy can stabilize the progression of CF lung disease has recently been established in a Phase IIb trial. An early phase study to assess the safety and explore efficacy of CFTR mRNA repair is ongoing, while mRNA delivery and genome editing-based strategies are currently at the pre-clinical phase of development. This review has been written jointly by some of those involved in the various CF "genetic medicine" fields and will summarize the current state-of-the-art, as well as discuss future developments. Where applicable, it highlights common problems faced by each of the strategies, and also tries to highlight where a specific strategy may have an advantage on the pathway to clinical translation. We hope that this review will contribute to the ongoing discussion about the hype versus reality of genetic medicine-based treatment approaches in CF. Pediatr Pulmonol. 2016;51:S5-S17. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eric W F W Alton
- UK Cystic Fibrosis Gene Therapy Consortium, Edinburgh, Oxford, London
| | | | - Jane C Davies
- UK Cystic Fibrosis Gene Therapy Consortium, Edinburgh, Oxford, London
| | - Deborah R Gill
- UK Cystic Fibrosis Gene Therapy Consortium, Edinburgh, Oxford, London
| | - Uta Griesenbach
- UK Cystic Fibrosis Gene Therapy Consortium, Edinburgh, Oxford, London.
| | - Patrick T Harrison
- Department of Physiology and BioSciences Institute, University College Cork, Cork, Ireland
| | | | - Tracy Higgins
- UK Cystic Fibrosis Gene Therapy Consortium, Edinburgh, Oxford, London
| | - Stephen C Hyde
- UK Cystic Fibrosis Gene Therapy Consortium, Edinburgh, Oxford, London
| | - J Alastair Innes
- UK Cystic Fibrosis Gene Therapy Consortium, Edinburgh, Oxford, London
| | - Michael S D Korman
- Department of Pediatrics I - Pediatric Infectiology and Immunology - Translational Genomics and Gene Therapy, University of Tübingen, Tübingen, Germany
| |
Collapse
|
35
|
Amrollahi P, Shah B, Seifi A, Tayebi L. Recent advancements in regenerative dentistry: A review. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:1383-90. [PMID: 27612840 DOI: 10.1016/j.msec.2016.08.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 08/04/2016] [Accepted: 08/18/2016] [Indexed: 12/20/2022]
Abstract
Although human mouth benefits from remarkable mechanical properties, it is very susceptible to traumatic damages, exposure to microbial attacks, and congenital maladies. Since the human dentition plays a crucial role in mastication, phonation and esthetics, finding promising and more efficient strategies to reestablish its functionality in the event of disruption has been important. Dating back to antiquity, conventional dentistry has been offering evacuation, restoration, and replacement of the diseased dental tissue. However, due to the limited ability and short lifespan of traditional restorative solutions, scientists have taken advantage of current advancements in medicine to create better solutions for the oral health field and have coined it "regenerative dentistry." This new field takes advantage of the recent innovations in stem cell research, cellular and molecular biology, tissue engineering, and materials science etc. In this review, the recently known resources and approaches used for regeneration of dental and oral tissues were evaluated using the databases of Scopus and Web of Science. Scientists have used a wide range of biomaterials and scaffolds (artificial and natural), genes (with viral and non-viral vectors), stem cells (isolated from deciduous teeth, dental pulp, periodontal ligament, adipose tissue, salivary glands, and dental follicle) and growth factors (used for stimulating cell differentiation) in order to apply tissue engineering approaches to dentistry. Although they have been successful in preclinical and clinical partial regeneration of dental tissues, whole-tooth engineering still seems to be far-fetched, unless certain shortcomings are addressed.
Collapse
Affiliation(s)
- Pouya Amrollahi
- Helmerich Advanced Technology Research Center, School of Material Science and Engineering, Oklahoma State University, Tulsa, OK 74106, USA
| | - Brinda Shah
- Marquette University School of Dentistry, Milwaukee, WI 53201, USA
| | - Amir Seifi
- Marquette University School of Dentistry, Milwaukee, WI 53201, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53201, USA; Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK.
| |
Collapse
|
36
|
Li Y, Hei M, Xu Y, Qian X, Zhu W. Ammonium salt modified mesoporous silica nanoparticles for dual intracellular-responsive gene delivery. Int J Pharm 2016; 511:689-702. [PMID: 27426108 DOI: 10.1016/j.ijpharm.2016.07.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 06/30/2016] [Accepted: 07/14/2016] [Indexed: 01/12/2023]
Abstract
Effective gene delivery system plays an importmant role in the gene therapy. Mesoporous silica nanoparticle (MSN) has become one potential gene delivery vector because of its high stability, good biodegradability and low cytotoxicity. Herein, MSN-based dual intracellular responsive gene delivery system CMSN-A was designed and fabricated. Short chain ammonium group, which is modified with disulfide bond and amide bond simultaneously, is facilely grafted onto the mesoporous silica nanoparticles. As-synthesized CMSN-A is endowed with small size (80-110nm), large conical pores (15-23nm), and moderate Zeta potential (+25±2mV), which behaves high gene loading capacity, good stability and effectively gene transfection. Moreover, CMSN-A exhibits dual micro-environment responsive (lower pH, more reducing substances) due to the redox-sensitive disulfide bond and pH-sensitive amide bond in the short chain ammonium group. The cellular uptake study indicates that CMSN-A could transfer both plasmid DNA (pDNA) and siRNA into different kinds of tumour cells, which demonstrate the promising potential of CMSN-A as effective and safe gene-delivery vectors.
Collapse
Affiliation(s)
- Yujie Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Mingyang Hei
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yufang Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xuhong Qian
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weiping Zhu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
37
|
Kim N, Duncan GA, Hanes J, Suk JS. Barriers to inhaled gene therapy of obstructive lung diseases: A review. J Control Release 2016; 240:465-488. [PMID: 27196742 DOI: 10.1016/j.jconrel.2016.05.031] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 12/29/2022]
Abstract
Knowledge of genetic origins of obstructive lung diseases has made inhaled gene therapy an attractive alternative to the current standards of care that are limited to managing disease symptoms. Initial lung gene therapy clinical trials occurred in the early 1990s following the discovery of the genetic defect responsible for cystic fibrosis (CF), a monogenic disorder. However, despite over two decades of intensive effort, gene therapy has yet to help patients with CF or any other obstructive lung disease. The slow progress is due in part to poor understanding of the biological barriers to inhaled gene therapy. Encouragingly, clinical trials have shown that inhaled gene therapy with various viral vectors and non-viral gene vectors is well tolerated by patients, and continued research has provided valuable lessons and resources that may lead to future success of this therapeutic strategy. In this review, we first introduce representative obstructive lung diseases and examine limitations of currently available therapeutic options. We then review key components for successful execution of inhaled gene therapy, including gene delivery systems, primary physiological barriers and strategies to overcome them, and advances in preclinical disease models with which the most promising systems may be identified for human clinical trials.
Collapse
Affiliation(s)
- Namho Kim
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gregg A Duncan
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Justin Hanes
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Environmental and Health Sciences, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jung Soo Suk
- The Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
38
|
Teagle AR, Birchall JC, Hargest R. Gene Therapy for Pyoderma Gangrenosum: Optimal Transfection Conditions and Effect of Drugs on Gene Delivery in the HaCaT Cell Line Using Cationic Liposomes. Skin Pharmacol Physiol 2016; 29:119-29. [DOI: 10.1159/000444859] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/19/2016] [Indexed: 11/19/2022]
|
39
|
Paul-Smith MC, Bell RV, Alton WE, Alton EW, Griesenbach U. Gene therapy for cystic fibrosis: recent progress and current aims. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1180974] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Michael C. Paul-Smith
- Department of Gene Therapy and the UK Cystic Fibrosis Gene Therapy Consortium, Imperial College, London, UK
| | - Robyn V. Bell
- Department of Gene Therapy and the UK Cystic Fibrosis Gene Therapy Consortium, Imperial College, London, UK
| | - William E. Alton
- Department of Gene Therapy and the UK Cystic Fibrosis Gene Therapy Consortium, Imperial College, London, UK
| | - Eric W.F.W. Alton
- Department of Gene Therapy and the UK Cystic Fibrosis Gene Therapy Consortium, Imperial College, London, UK
| | - Uta Griesenbach
- Department of Gene Therapy and the UK Cystic Fibrosis Gene Therapy Consortium, Imperial College, London, UK
| |
Collapse
|
40
|
Youngren-Ortiz SR, Gandhi NS, España-Serrano L, Chougule MB. Aerosol Delivery of siRNA to the Lungs. Part 2: Nanocarrier-based Delivery Systems. KONA : POWDER SCIENCE AND TECHNOLOGY IN JAPAN 2016; 34:44-69. [PMID: 28392618 PMCID: PMC5381822 DOI: 10.14356/kona.2017005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
In this article, applications of engineered nanoparticles containing siRNA for inhalation delivery are reviewed and discussed. Diseases with identified protein malfunctions may be mitigated through the use of well-designed siRNA therapeutics. The inhalation route of administration provides local delivery of siRNA therapeutics to the lungs for various pulmonary diseases. A siRNA delivery system can be used to overcome the barriers of pulmonary delivery, such as anatomical barriers, mucociliary clearance, cough clearance, and alveolar macrophage clearance. Apart from naked siRNA aerosol delivery, previously studied siRNA carrier systems include those of lipidic, polymeric, peptide, or inorganic origin. These delivery systems can achieve pulmonary delivery through the generation of an aerosol via an inhaler or nebulizer. The preparation methodologies for these siRNA nanocarrier systems will be discussed herein. The use of inhalable nanocarrier siRNA delivery systems have barriers to their effective delivery, but overcoming these constraints while formulating a safe and effective delivery system will offer unique advances to the field of inhaled medicine.
Collapse
Affiliation(s)
- Susanne R. Youngren-Ortiz
- Translational Drug Delivery Research (TransDDR) Laboratory, Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, USA
| | - Nishant S. Gandhi
- Translational Drug Delivery Research (TransDDR) Laboratory, Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, USA
| | - Laura España-Serrano
- Translational Drug Delivery Research (TransDDR) Laboratory, Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, USA
| | - Mahavir B. Chougule
- Translational Drug Delivery Research (TransDDR) Laboratory, Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, USA
- Natural Products and Experimental Therapeutics Program, The Cancer Research Center, University of Hawaii at Manoa, Honolulu, Hawaii 96813, USA
| |
Collapse
|
41
|
Bazzani RP, Pringle IA, Connolly MM, Davies LA, Sumner-Jones SG, Schleef M, Hyde SC, Gill DR. Transgene sequences free of CG dinucleotides lead to high level, long-term expression in the lung independent of plasmid backbone design. Biomaterials 2016; 93:20-26. [PMID: 27061267 DOI: 10.1016/j.biomaterials.2016.03.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/17/2016] [Accepted: 03/17/2016] [Indexed: 12/13/2022]
Abstract
Non-viral aerosol gene therapy offers great potential for treating chronic lung diseases of the airways such as cystic fibrosis (CF). Early clinical trials showed that transgene expression in the airways was transient whereas maximal duration of transgene expression is essential in order to minimise the frequency of aerosol treatments. Improved vector design, such as careful selection of the promoter/enhancer, can lead to more persistent levels of transgene expression, but multiple factors affect expression in vivo. Following aerosol delivery to the lungs of mice, we measured reporter gene expression from a CpG-free luciferase transgene cassette in the context of both a plasmid and minicircle vector configuration and showed that the vector backbone had no effect on expression. Transgene activity was affected by the vector backbone however, when a similar, but sub-optimal CpG-containing transgene was used, suggesting that aspects of the plasmid backbone had a negative impact on transgene expression. Similar studies were performed in Toll-like receptor-9 (TLR9) knockout mice to investigate a potential role for the TLR9 signalling pathway in detecting CpGs in the vector sequence. Even in the absence of TLR9, persistent expression could only be achieved with a CpG-free transgene. Together, these data indicate that in order to achieve high levels of persistent expression in vivo, a CpG-free transgene cassette is required.
Collapse
Affiliation(s)
- Reto P Bazzani
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK; The UK Cystic Fibrosis Gene Therapy Consortium, UK
| | - Ian A Pringle
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK; The UK Cystic Fibrosis Gene Therapy Consortium, UK
| | - Mary M Connolly
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK; The UK Cystic Fibrosis Gene Therapy Consortium, UK
| | - Lee A Davies
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK; The UK Cystic Fibrosis Gene Therapy Consortium, UK
| | - Stephanie G Sumner-Jones
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK; The UK Cystic Fibrosis Gene Therapy Consortium, UK
| | - Martin Schleef
- PlasmidFactory, Meisenstraße 96, D-33607 Bielefeld, Germany
| | - Stephen C Hyde
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK; The UK Cystic Fibrosis Gene Therapy Consortium, UK
| | - Deborah R Gill
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, OX3 9DU, UK; The UK Cystic Fibrosis Gene Therapy Consortium, UK.
| |
Collapse
|
42
|
Dhooghe B, Haaf JB, Noel S, Leal T. Strategies in early clinical development for the treatment of basic defects of cystic fibrosis. Expert Opin Investig Drugs 2016; 25:423-36. [PMID: 26878157 DOI: 10.1517/13543784.2016.1154041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Twenty-six years after the identification of the gene responsible for cystic fibrosis (CF), controversies still surround the pathogenesis of the disease that continues to burden and shorten lives. Therefore, finding effective therapeutic strategies that target the basic defect of CF is crucially needed. AREAS COVERED This review offers a comprehensive survey of fundamental therapies in early stages of development for the treatment of CF. The first part describes recent strategies targeting the basic defect either at the gene or at the transcript level. The second part summarizes a panel of novel strategies targeting protein repair. The third part reports strategies targeting non-CFTR channels. EXPERT OPINION Recent major breakthroughs in CF therapy have been made, raising hope to find a cure for CF. Apart from Vertex corrector and potentiator molecules (lumacaftor, ivacaftor, VX-661) and from ataluren, used to correct nonsense mutations, most compounds being currently tested are in very early (I-II) phases of development and definitive clinical results are keenly expected. Among the broad list of molecules and strategies being tested, the QR-010 compound and inhibitors of phosphodiesterase type 5 (sildenafil, vardenafil) could reveal a strong potentiality as therapeutic candidates to cure CF.
Collapse
Affiliation(s)
- Barbara Dhooghe
- a Louvain centre for Toxicology and Applied Pharmacology , Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain , Brussels , Belgium
| | - Jérémy Boris Haaf
- a Louvain centre for Toxicology and Applied Pharmacology , Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain , Brussels , Belgium
| | - Sabrina Noel
- a Louvain centre for Toxicology and Applied Pharmacology , Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain , Brussels , Belgium
| | - Teresinha Leal
- a Louvain centre for Toxicology and Applied Pharmacology , Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain , Brussels , Belgium
| |
Collapse
|
43
|
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) gene was identified in 1989. This opened the door for the development of cystic fibrosis (CF) gene therapy, which has been actively pursued for the last 20 years. Although 26 clinical trials involving approximately 450 patients have been carried out, the vast majority of these trials were short and included small numbers of patients; they were not designed to assess clinical benefit, but to establish safety and proof-of-concept for gene transfer using molecular end points such as the detection of recombinant mRNA or correction of the ion transport defect. The only currently published trial designed and powered to assess clinical efficacy (defined as improvement in lung function) administered AAV2-CFTR to the lungs of patients with CF. The U.K. Cystic Fibrosis Gene Therapy Consortium completed, in the autumn of 2014, the first nonviral gene therapy trial designed to answer whether repeated nonviral gene transfer (12 doses over 12 months) can lead to clinical benefit. The demonstration that the molecular defect in CFTR can be corrected with small-molecule drugs, and the success of gene therapy in other monogenic diseases, is boosting interest in CF gene therapy. Developments are discussed here.
Collapse
Affiliation(s)
- Uta Griesenbach
- Department of Gene Therapy and the U.K. Cystic Fibrosis Gene Therapy Consortium, Imperial College, London SW3 6LR, United Kingdom
| | - Kamila M Pytel
- Department of Gene Therapy and the U.K. Cystic Fibrosis Gene Therapy Consortium, Imperial College, London SW3 6LR, United Kingdom
| | - Eric W F W Alton
- Department of Gene Therapy and the U.K. Cystic Fibrosis Gene Therapy Consortium, Imperial College, London SW3 6LR, United Kingdom
| |
Collapse
|
44
|
André EM, Pensado A, Resnier P, Braz L, Rosa da Costa AM, Passirani C, Sanchez A, Montero-Menei CN. Characterization and comparison of two novel nanosystems associated with siRNA for cellular therapy. Int J Pharm 2015; 497:255-67. [PMID: 26617318 DOI: 10.1016/j.ijpharm.2015.11.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/08/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022]
Abstract
To direct stem cell fate, a delicate control of gene expression through small interference RNA (siRNA) is emerging as a new and safe promising strategy. In this way, the expression of proteins hindering neuronal commitment may be transiently inhibited thus driving differentiation. Mesenchymal stem cells (MSC), which secrete tissue repair factors, possess immunomodulatory properties and may differentiate towards the neuronal lineage, are a promising cell source for cell therapy studies in the central nervous system. To better drive their neuronal commitment the repressor Element-1 silencing transcription (REST) factor, may be inhibited by siRNA technology. The design of novel nanoparticles (NP) capable of safely delivering nucleic acids is crucial in order to successfully develop this strategy. In this study we developed and characterized two different siRNA NP. On one hand, sorbitan monooleate (Span(®)80) based NP incorporating the cationic components poly-l-arginine or cationized pullulan, thus allowing the association of siRNA were designed. These NP presented a small size (205 nm) and a negative surface charge (-38 mV). On the other hand, lipid nanocapsules (LNC) associating polymers with lipids and allowing encapsulation of siRNA complexed with lipoplexes were also developed. Their size was of 82 nm with a positive surface charge of +7 mV. Both NP could be frozen with appropriate cryoprotectors. Cytotoxicity and transfection efficiency at different siRNA doses were monitored by evaluating REST expression. An inhibition of around 60% of REST expression was observed with both NP when associating 250 ng/mL of siRNA-REST, as recommended for commercial reagents. Span NP were less toxic for human MSCs than LNCs, but although both NP showed a similar inhibition of REST over time and the induction of neuronal commitment, LNC-siREST induced a higher expression of neuronal markers. Therefore, two different tailored siRNA NP offering great potential for human stem cell differentiation have been developed, encouraging the pursuit of further in vitro and in vivo in studies.
Collapse
Affiliation(s)
- E M André
- PRES LUNAM-University of Angers, F-49933 Angers, France; INSERM U1066-Micro et Nanomédecines Biomimétiques, 4 rue larrey, F-49933 Angers, France
| | - A Pensado
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, Campus Vida, 15782 Santiago de Compostela, Spain
| | - P Resnier
- PRES LUNAM-University of Angers, F-49933 Angers, France; INSERM U1066-Micro et Nanomédecines Biomimétiques, 4 rue larrey, F-49933 Angers, France
| | - L Braz
- CIQA-Algarve Chemistry Research Centre, University of Algarve, 8005-139 Faro, Portugal; School of Health-University of Algarve, 8000-510 Faro, Portugal; Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - A M Rosa da Costa
- CIQA-Algarve Chemistry Research Centre, University of Algarve, 8005-139 Faro, Portugal; Department of Chemistry and Pharmacy, Faculty of Science and Technology, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - C Passirani
- PRES LUNAM-University of Angers, F-49933 Angers, France; INSERM U1066-Micro et Nanomédecines Biomimétiques, 4 rue larrey, F-49933 Angers, France
| | - A Sanchez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, Campus Vida, 15782 Santiago de Compostela, Spain; Molecular Image Group. Health Research Institute-University Clinical Hospital of Santiago de Compostela (IDIS), A Choupana, 15706 Santiago de Compostela, Spain
| | - C N Montero-Menei
- PRES LUNAM-University of Angers, F-49933 Angers, France; INSERM U1066-Micro et Nanomédecines Biomimétiques, 4 rue larrey, F-49933 Angers, France.
| |
Collapse
|
45
|
Singh Y, Tomar S, Khan S, Meher JG, Pawar VK, Raval K, Sharma K, Singh PK, Chaurasia M, Surendar Reddy B, Chourasia MK. Bridging small interfering RNA with giant therapeutic outcomes using nanometric liposomes. J Control Release 2015; 220:368-387. [PMID: 26528900 DOI: 10.1016/j.jconrel.2015.10.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 01/04/2023]
Abstract
The scope of RNAi based therapeutics is unquestionable. However, if we dissect the current trend of clinical trials for afore mentioned drug class, some stark trends appear: 1) naked siRNA only exerts influence in topical mode whilst systemic delivery requires a carrier and 2) even after two decades of extensive efforts, not even a single siRNA containing product is commercially available. It was therefore felt that a perspective simplifying the unique intricacies of working with a merger of siRNA and liposomes from a pharmaceutical viewpoint could draw the attention of a wider array of interested researchers. We begin from the beginning and attempt to conduit the gap between theoretical logic and experimental/actual constraints. This, in turn could stimulate the next generation of investigators, gearing them to tackle the conundrum, which is siRNA delivery.
Collapse
Affiliation(s)
- Yuvraj Singh
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sandeep Tomar
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shariq Khan
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Jaya Gopal Meher
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Vivek K Pawar
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Kavit Raval
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Komal Sharma
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pankaj K Singh
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Mohini Chaurasia
- Amity Institute of Pharmacy, Amity University, Lucknow, UP 226028, India
| | - B Surendar Reddy
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Manish K Chourasia
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
46
|
Smith RJ, Beck RW, Prevette LE. Impact of molecular weight and degree of conjugation on the thermodynamics of DNA complexation and stability of polyethylenimine-graft-poly(ethylene glycol) copolymers. Biophys Chem 2015; 203-204:12-21. [DOI: 10.1016/j.bpc.2015.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 12/13/2022]
|
47
|
Yan Z, Stewart ZA, Sinn PL, Olsen JC, Hu J, McCray PB, Engelhardt JF. Ferret and pig models of cystic fibrosis: prospects and promise for gene therapy. HUM GENE THER CL DEV 2015; 26:38-49. [PMID: 25675143 PMCID: PMC4367511 DOI: 10.1089/humc.2014.154] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 12/26/2014] [Indexed: 12/13/2022] Open
Abstract
Large animal models of genetic diseases are rapidly becoming integral to biomedical research as technologies to manipulate the mammalian genome improve. The creation of cystic fibrosis (CF) ferrets and pigs is an example of such progress in animal modeling, with the disease phenotypes in the ferret and pig models more reflective of human CF disease than mouse models. The ferret and pig CF models also provide unique opportunities to develop and assess the effectiveness of gene and cell therapies to treat affected organs. In this review, we examine the organ disease phenotypes in these new CF models and the opportunities to test gene therapies at various stages of disease progression in affected organs. We then discuss the progress in developing recombinant replication-defective adenoviral, adeno-associated viral, and lentiviral vectors to target genes to the lung and pancreas in ferrets and pigs, the two most affected organs in CF. Through this review, we hope to convey the potential of these new animal models for developing CF gene and cell therapies.
Collapse
Affiliation(s)
- Ziying Yan
- Department of Anatomy and Cell Biology, University of Iowa School of Medicine, Iowa City, IA 52242
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
| | - Zoe A. Stewart
- Department of Surgery, University of Iowa School of Medicine, Iowa City, IA 52242
| | - Patrick L. Sinn
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Pediatrics, University of Iowa School of Medicine, Iowa City, IA 52242
| | - John C. Olsen
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jim Hu
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children and University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Paul B. McCray
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Pediatrics, University of Iowa School of Medicine, Iowa City, IA 52242
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa School of Medicine, Iowa City, IA 52242
- Center for Gene Therapy of Cystic Fibrosis, University of Iowa School of Medicine, Iowa City, IA 52242
- Department of Internal Medicine, University of Iowa School of Medicine, Iowa City, IA 52242
| |
Collapse
|
48
|
Gupta K, Singh S, Garg KN. Gene therapy in dentistry: Tool of genetic engineering. Revisited. Arch Oral Biol 2015; 60:439-46. [DOI: 10.1016/j.archoralbio.2014.11.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 11/26/2014] [Accepted: 11/27/2014] [Indexed: 01/17/2023]
|
49
|
Synthesis and in vitro transfection efficiency of spermine-based cationic lipids with different central core structures and lipophilic tails. Bioorg Med Chem Lett 2015; 25:496-503. [DOI: 10.1016/j.bmcl.2014.12.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 11/24/2014] [Accepted: 12/12/2014] [Indexed: 01/17/2023]
|
50
|
Dan N. Lipid-Nucleic Acid Supramolecular Complexes: Lipoplex Structure and the Kinetics of Formation. AIMS BIOPHYSICS 2015. [DOI: 10.3934/biophy.2015.2.163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|