1
|
Antony JS, Birrer P, Bohnert C, Zimmerli S, Hillmann P, Schaffhauser H, Hoeflich C, Hoeflich A, Khairallah R, Satoh AT, Kappeler I, Ferreira I, Zuideveld KP, Metzger F. Local application of engineered insulin-like growth factor I mRNA demonstrates regenerative therapeutic potential in vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102055. [PMID: 37928443 PMCID: PMC10622308 DOI: 10.1016/j.omtn.2023.102055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023]
Abstract
Insulin-like growth factor I (IGF-I) is a growth-promoting anabolic hormone that fosters cell growth and tissue homeostasis. IGF-I deficiency is associated with several diseases, including growth disorders and neurological and musculoskeletal diseases due to impaired regeneration. Despite the vast regenerative potential of IGF-I, its unfavorable pharmacokinetic profile has prevented it from being used therapeutically. In this study, we resolved these challenges by the local administration of IGF-I mRNA, which ensures desirable homeostatic kinetics and non-systemic, local dose-dependent expression of IGF-I protein. Furthermore, IGF-I mRNA constructs were sequence engineered with heterologous signal peptides, which improved in vitro protein secretion (2- to 6-fold) and accelerated in vivo functional regeneration (16-fold) over endogenous IGF-I mRNA. The regenerative potential of engineered IGF-I mRNA was validated in a mouse myotoxic muscle injury and rabbit spinal disc herniation models. Engineered IGF-I mRNA had a half-life of 17-25 h in muscle tissue and showed dose-dependent expression of IGF-I over 2-3 days. Animal models confirm that locally administered IGF-I mRNA remained at the site of injection, contributing to the safety profile of mRNA-based treatment in regenerative medicine. In summary, we demonstrate that engineered IGF-I mRNA holds therapeutic potential with high clinical translatability in different diseases.
Collapse
Affiliation(s)
| | | | | | - Sina Zimmerli
- Versameb AG, Technology Park, 4057 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Miao H, Huang K, Li Y, Li R, Zhou X, Shi J, Tong Z, Sun Z, Yu A. Optimization of formulation and atomization of lipid nanoparticles for the inhalation of mRNA. Int J Pharm 2023; 640:123050. [PMID: 37201764 DOI: 10.1016/j.ijpharm.2023.123050] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/06/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023]
Abstract
Lipid nanoparticles (LNPs) have demonstrated efficacy and safety for mRNA vaccine administration by intramuscular injection; however, the pulmonary delivery of mRNA encapsulated LNPs remains challenging. The atomization process of LNPs will cause shear stress due to dispersed air, air jets, ultrasonication, vibrating mesh etc., leading to the agglomeration or leakage of LNPs, which can be detrimental to transcellular transport and endosomal escape. In this study, the LNP formulation, atomization methods and buffer system were optimized to maintain the LNP stability and mRNA efficiency during the atomization process. Firstly, a suitable LNP formulation for atomization was optimized based on the in vitro results, and the optimized LNP formulation was AX4, DSPC, cholesterol and DMG-PEG2K at a 35/16/46.5/2.5 (%) molar ratio. Subsequently, different atomization methods were compared to find the most suitable method to deliver mRNA-LNP solution. Soft mist inhaler (SMI) was found to be the best for pulmonary delivery of mRNA encapsulated LNPs. The physico-chemical properties such as size and entrapment efficiency (EE) of the LNPs were further improved by adjusting the buffer system with trehalose. Lastly, the in vivo fluorescence imaging of mice demonstrated that SMI with proper LNPs design and buffer system hold promise for inhaled mRNA-LNP therapies.
Collapse
Affiliation(s)
- Hao Miao
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia; Monash Suzhou Research Institute, Suzhou, 215000, China
| | - Ke Huang
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia; Monash Suzhou Research Institute, Suzhou, 215000, China
| | - Yingwen Li
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou, 215000, China
| | - Renjie Li
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia; Monash Suzhou Research Institute, Suzhou, 215000, China
| | - Xudong Zhou
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia; Monash Suzhou Research Institute, Suzhou, 215000, China
| | - Jingyu Shi
- School of Energy and Environment, Southeast University, Nanjing, 210000, China; Southeast University-Monash University Joint Research Institute, Suzhou, 215000 China
| | - Zhenbo Tong
- School of Energy and Environment, Southeast University, Nanjing, 210000, China; Southeast University-Monash University Joint Research Institute, Suzhou, 215000 China
| | - Zhenhua Sun
- Suzhou CureMed Biopharma Technology Co., Ltd., Suzhou, 215000, China.
| | - Aibing Yu
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia; Southeast University-Monash University Joint Research Institute, Suzhou, 215000 China
| |
Collapse
|
3
|
Moskowitzova K, Whitlock AE, Zurakowski D, Fauza DO. Hematogenous Routing of Exogenous mRNA Delivered Into the Amniotic Fluid. J Surg Res 2023; 289:116-120. [PMID: 37104922 DOI: 10.1016/j.jss.2023.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/21/2023] [Accepted: 03/25/2023] [Indexed: 04/29/2023]
Abstract
INTRODUCTION Therapies based on exogenous messenger RNA (mRNA) administration have emerged as a powerful novel strategy for the actual or potential treatment of an assortment of diseases, including congenital surgical pathologies. We sought to determine whether the minimally invasive transamniotic route could be an alternative for prenatal mRNA delivery. METHODS Pregnant Sprague-Dawley dams underwent laparotomy followed by volume-matched intra-amniotic injections in all their fetuses (n = 120) of either a suspension of a custom firefly luciferase mRNA encapsulated by a lipid- and synthetic cationic polymer-based composite, or of a suspension of the same encapsulation components without mRNA, on gestational day 17 (E17; term = E21-22). On E18, E19, E20, and E21, samples from 14 fetal anatomical sites and maternal serum were procured for the screening of mRNA incorporation by host cells by measurement of luciferase activity via microplate luminometry. Statistical analysis was by Mann-Whitney U-test, including Bonferroni-adjustment. RESULTS Overall survival was 87.5% (105/120). Controlled by the encapsulating composite without mRNA, luciferase activity was detected in the animals that received encapsulated mRNA in the following fetal annexes: amniotic fluid, amnion, chorion, umbilical cord, and placenta (P = 0.033 to <0.001), as well as in the following fetal sites: liver, stomach, intestines, and lungs (P = 0.043-0.002). CONCLUSIONS Packaged exogenous mRNA can be incorporated by the fetus at least at select anatomical sites after simple intra-amniotic administration in a rodent model. The pattern and chronology of mRNA incorporation are compatible with transplacental hematogenous routing, as well as with fetal swallowing/aspiration. Further study of transamniotic mRNA administration is warranted.
Collapse
Affiliation(s)
- Kamila Moskowitzova
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, Massachusetts
| | - Ashlyn E Whitlock
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, Massachusetts
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, Massachusetts
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
4
|
Haque AKMA, Weinmann P, Biswas S, Handgretinger R, Mezger M, Kormann MSD, Antony JS. RNA ImmunoGenic Assay: A Method to Detect Immunogenicity of in vitro Transcribed mRNA in Human Whole Blood. Bio Protoc 2020; 10:e3850. [PMID: 33855105 PMCID: PMC8032499 DOI: 10.21769/bioprotoc.3850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 11/02/2022] Open
Abstract
The mRNA therapeutics is a new class of medicine to treat many various diseases. However, in vitro transcribed (IVT) mRNA triggers immune responses due to recognition by human endosomal and cytoplasmic RNA sensors, but incorporation of modified nucleosides have been shown to reduce such responses. Therefore, an assay signifying important aspects of the human immune system is still required. Here, we present a simple ex vivo method called 'RNA ImmunoGenic Assay' to measure immunogenicity of IVT-mRNAs in human whole blood. Chemically modified and unmodified mRNA are complexed with a transfection reagent (TransIT), and co-incubated in human whole blood. Specific cytokines are measured (TNF-α, INF-α, INF-γ, IL-6 and IL-12p70) using ELISAs. The qPCR analysis is performed to reveal the activation of specific immune pathways. The RNA ImmunoGenic Assay provides a simple and fast method to detect donor specific - immune response against mRNA therapeutics. Graphic abstract: Schematic representation of RNA ImmunoGenic Assay.
Collapse
Affiliation(s)
- AKM Ashiqul Haque
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Germany
| | - Petra Weinmann
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Germany
| | - Sumit Biswas
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Germany
| | - Rupert Handgretinger
- University Children's Clinic Department of Paediatrics I, Hematology and Oncology, University of Tuebingen, Germany
| | - Markus Mezger
- University Children's Clinic Department of Paediatrics I, Hematology and Oncology, University of Tuebingen, Germany
| | - Michael S. D. Kormann
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Germany
| | - Justin S Antony
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Germany
- University Children's Clinic Department of Paediatrics I, Hematology and Oncology, University of Tuebingen, Germany
| |
Collapse
|
5
|
Zhang H, Leal J, Soto MR, Smyth HDC, Ghosh D. Aerosolizable Lipid Nanoparticles for Pulmonary Delivery of mRNA through Design of Experiments. Pharmaceutics 2020; 12:E1042. [PMID: 33143328 PMCID: PMC7692784 DOI: 10.3390/pharmaceutics12111042] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/24/2020] [Accepted: 10/28/2020] [Indexed: 01/07/2023] Open
Abstract
Messenger RNA is a class of promising nucleic acid therapeutics to treat a variety of diseases, including genetic diseases. The development of a stable and efficacious mRNA pulmonary delivery system would enable high therapeutic concentrations locally in the lungs to improve efficacy and limit potential toxicities. In this study, we employed a Design of Experiments (DOE) strategy to screen a library of lipid nanoparticle compositions to identify formulations possessing high potency both before and after aerosolization. Lipid nanoparticles (LNPs) showed stable physicochemical properties for at least 14 days of storage at 4 °C, and most formulations exhibited high encapsulation efficiencies greater than 80%. Generally, upon nebulization, LNP formulations showed increased particle size and decreased encapsulation efficiencies. An increasing molar ratio of poly-(ethylene) glycol (PEG)-lipid significantly decreased size but also intracellular protein expression of mRNA. We identified four formulations possessing higher intracellular protein expression ability in vitro even after aerosolization which were then assessed in in vivo studies. It was found that luciferase protein was predominately expressed in the mouse lung for the four lead formulations before and after nebulization. This study demonstrated that LNPs hold promise to be applied for aerosolization-mediated pulmonary mRNA delivery.
Collapse
Affiliation(s)
| | | | | | | | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (H.Z.); (J.L.); (M.R.S.); (H.D.C.S.)
| |
Collapse
|
6
|
DeRosa F, Smith L, Shen Y, Huang Y, Pan J, Xie H, Yahalom B, Heartlein MW. Improved Efficacy in a Fabry Disease Model Using a Systemic mRNA Liver Depot System as Compared to Enzyme Replacement Therapy. Mol Ther 2019; 27:878-889. [PMID: 30879951 PMCID: PMC6453518 DOI: 10.1016/j.ymthe.2019.03.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/30/2022] Open
Abstract
Fabry disease is a lysosomal storage disorder caused by the deficiency of α-galactosidase A. Enzyme deficiency results in a progressive decline in renal and cardiac function, leading to cardiomyopathy and end-stage renal disease. Current treatments available, including enzyme replacement therapies, have provided significant benefit to patients; however, unmet medical needs remain. mRNA therapy, with drug-like properties, has the unique ability to produce therapeutic proteins endogenously. Here we describe the sustained delivery of therapeutic human α-galactosidase protein in vivo via nanoparticle-formulated mRNA in mouse and non-human primate, with a demonstration of efficacy through clinically relevant biomarker reduction in a mouse Fabry disease model. Multi-component nanoparticles formulated with lipids and lipid-like materials were developed for the delivery of mRNA encoding human α-galactosidase protein. Upon delivery of human GLA mRNA to mice, serum GLA protein levels reached as high as ∼1,330-fold over normal physiological values.
Collapse
Affiliation(s)
| | | | | | - Yan Huang
- Shire Pharmaceuticals, Lexington, MA 02141, USA
| | - Jing Pan
- Shire Pharmaceuticals, Lexington, MA 02141, USA
| | | | | | | |
Collapse
|
7
|
Recent Developments in mRNA-Based Protein Supplementation Therapy to Target Lung Diseases. Mol Ther 2019; 27:803-823. [PMID: 30905577 DOI: 10.1016/j.ymthe.2019.02.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022] Open
Abstract
Protein supplementation therapy using in vitro-transcribed (IVT) mRNA for genetic diseases contains huge potential as a new class of therapy. From the early ages of synthetic mRNA discovery, a great number of studies showed the versatile use of IVT mRNA as a novel approach to supplement faulty or absent protein and also as a vaccine. Many modifications have been made to produce high expressions of mRNA causing less immunogenicity and more stability. Recent advancements in the in vivo lung delivery of mRNA complexed with various carriers encouraged the whole mRNA community to tackle various genetic lung diseases. This review gives a comprehensive overview of cells associated with various lung diseases and recent advancements in mRNA-based protein replacement therapy. This review also covers a brief summary of developments in mRNA modifications and nanocarriers toward clinical translation.
Collapse
|
8
|
Grassin-Delyle S, Abrial C, Salvator H, Brollo M, Naline E, Devillier P. The Role of Toll-Like Receptors in the Production of Cytokines by Human Lung Macrophages. J Innate Immun 2018; 12:63-73. [PMID: 30557876 DOI: 10.1159/000494463] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/13/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The Toll-like receptor (TLR) family is involved in the recognition of and response to microbial infections. These receptors are expressed in leukocytes. TLR stimulation induces the production of proinflammatory cytokines and chemokines. Given that human lung macrophages (LMs) constitute the first line of defense against inhaled pathogens, the objective of this study was to investigate the expression and function of TLR subtypes in this cell population. METHODS Human primary LMs were obtained from patients undergoing surgical resection. The RNA and protein expression levels of TLRs, chemokines, and cytokines were assessed after incubation with subtype-selective agonists. RESULTS In human LMs, the TLR expression level varied from one subtype to another. Stimulation with subtype-selective agonists induced an intense, concentration- and time-dependent increase in the production of chemokines and cytokines. TLR4 stimulation induced the strongest effect, whereas TLR9 stimulation induced a much weaker response. CONCLUSIONS The stimulation of TLRs in human LMs induces intense cytokine and chemokine production, a characteristic of the proinflammatory M1 macrophage phenotype.
Collapse
Affiliation(s)
- Stanislas Grassin-Delyle
- Département des Maladies Respiratoires, Hôpital Foch, Suresnes, France, .,INSERM UMR 1173 et Plateforme de spectrométrie de masse MasSpecLab, UFR des Sciences de la Santé Simone Veil, Université Versailles Saint Quentin, Université Paris Saclay, Montigny-le-Bretonneux, France,
| | - Charlotte Abrial
- Laboratoire de Pharmacologie UPRES EA220, Université Versailles Saint Quentin, Université Paris Saclay, Hôpital Foch, Suresnes, France
| | - Hélène Salvator
- Département des Maladies Respiratoires, Hôpital Foch, Suresnes, France.,Laboratoire de Pharmacologie UPRES EA220, Université Versailles Saint Quentin, Université Paris Saclay, Hôpital Foch, Suresnes, France
| | - Marion Brollo
- Laboratoire de Pharmacologie UPRES EA220, Université Versailles Saint Quentin, Université Paris Saclay, Hôpital Foch, Suresnes, France
| | - Emmanuel Naline
- Département des Maladies Respiratoires, Hôpital Foch, Suresnes, France.,Laboratoire de Pharmacologie UPRES EA220, Université Versailles Saint Quentin, Université Paris Saclay, Hôpital Foch, Suresnes, France
| | - Philippe Devillier
- Département des Maladies Respiratoires, Hôpital Foch, Suresnes, France.,Laboratoire de Pharmacologie UPRES EA220, Université Versailles Saint Quentin, Université Paris Saclay, Hôpital Foch, Suresnes, France
| |
Collapse
|
9
|
Antony JS, Latifi N, Haque AKMA, Lamsfus-Calle A, Daniel-Moreno A, Graeter S, Baskaran P, Weinmann P, Mezger M, Handgretinger R, Kormann MSD. Gene correction of HBB mutations in CD34 + hematopoietic stem cells using Cas9 mRNA and ssODN donors. Mol Cell Pediatr 2018; 5:9. [PMID: 30430274 PMCID: PMC6236008 DOI: 10.1186/s40348-018-0086-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 10/17/2018] [Indexed: 12/13/2022] Open
Abstract
Background β-Thalassemia is an inherited hematological disorder caused by mutations in the human hemoglobin beta (HBB) gene that reduce or abrogate β-globin expression. Although lentiviral-mediated expression of β-globin and autologous transplantation is a promising therapeutic approach, the risk of insertional mutagenesis or low transgene expression is apparent. However, targeted gene correction of HBB mutations with programmable nucleases such as CRISPR/Cas9, TALENs, and ZFNs with non-viral repair templates ensures a higher safety profile and endogenous expression control. Methods We have compared three different gene-editing tools (CRISPR/Cas9, TALENs, and ZFNs) for their targeting efficiency of the HBB gene locus. As a proof of concept, we studied the personalized gene-correction therapy for a common β-thalassemia splicing variant HBBIVS1–110 using Cas9 mRNA and several optimally designed single-stranded oligonucleotide (ssODN) donors in K562 and CD34+ hematopoietic stem cells (HSCs). Results Our results exhibited that indel frequency of CRISPR/Cas9 was superior to TALENs and ZFNs (P < 0.0001). Our designed sgRNA targeting the site of HBBIVS1–110 mutation showed indels in both K562 cells (up to 77%) and CD34+ hematopoietic stem cells—HSCs (up to 87%). The absolute quantification by next-generation sequencing showed that up to 8% site-specific insertion of the NheI tag was achieved using Cas9 mRNA and a chemically modified ssODN in CD34+ HSCs. Conclusion Our approach provides guidance on non-viral gene correction in CD34+ HSCs using Cas9 mRNA and chemically modified ssODN. However, further optimization is needed to increase the homology directed repair (HDR) to attain a real clinical benefit for β-thalassemia. Electronic supplementary material The online version of this article (10.1186/s40348-018-0086-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Justin S Antony
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany.,University Children's Hospital, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany.,Department of Hematology, Oncology, Clinical Immunology, University of Tuebingen, Tuebingen, Germany
| | - Ngadhnjim Latifi
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - A K M Ashiqul Haque
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - Andrés Lamsfus-Calle
- University Children's Hospital, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Alberto Daniel-Moreno
- University Children's Hospital, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Sebastian Graeter
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - Praveen Baskaran
- Center for Quantitative Biology, University of Tuebingen, Tuebingen, Germany
| | - Petra Weinmann
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - Markus Mezger
- University Children's Hospital, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Rupert Handgretinger
- University Children's Hospital, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Michael S D Kormann
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
10
|
Haque AKMA, Dewerth A, Antony JS, Riethmüller J, Schweizer GR, Weinmann P, Latifi N, Yasar H, Pedemonte N, Sondo E, Weidensee B, Ralhan A, Laval J, Schlegel P, Seitz C, Loretz B, Lehr CM, Handgretinger R, Kormann MSD. Chemically modified hCFTR mRNAs recuperate lung function in a mouse model of cystic fibrosis. Sci Rep 2018; 8:16776. [PMID: 30425265 PMCID: PMC6233194 DOI: 10.1038/s41598-018-34960-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022] Open
Abstract
Gene therapy has always been a promising therapeutic approach for Cystic Fibrosis (CF). However, numerous trials using DNA or viral vectors encoding the correct protein resulted in a general low efficacy. In the last years, chemically modified messenger RNA (cmRNA) has been proven to be a highly potent, pulmonary drug. Consequently, we first explored the expression, function and immunogenicity of human (h)CFTR encoded by cmRNAhCFTR in vitro and ex vivo, quantified the expression by flow cytometry, determined its function using a YFP based assay and checked the immune response in human whole blood. Similarly, we examined the function of cmRNAhCFTR in vivo after intratracheal (i.t.) or intravenous (i.v.) injection of the assembled cmRNAhCFTR together with Chitosan-coated PLGA (poly-D, L-lactide-co-glycolide 75:25 (Resomer RG 752 H)) nanoparticles (NPs) by FlexiVent. The amount of expression of human hCFTR encoded by cmRNAhCFTR was quantified by hCFTR ELISA, and cmRNAhCFTR values were assessed by RT-qPCR. Thereby, we observed a significant improvement of lung function, especially in regards to FEV0.1, suggesting NP-cmRNAhCFTR as promising therapeutic option for CF patients independent of their CFTR genotype.
Collapse
Affiliation(s)
- A K M Ashiqul Haque
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Alexander Dewerth
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Justin S Antony
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany.,Department of Hematology, Oncology, Clinical Immunology, University of Tuebingen, Tuebingen, Germany
| | | | - Georg R Schweizer
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Petra Weinmann
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Ngadhnjim Latifi
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Hanzey Yasar
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbruecken, Germany
| | | | - Elvira Sondo
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | - Brian Weidensee
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Anjali Ralhan
- Department of Pediatrics I - Immunology and Pneumology/Cystic fibrosis, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Julie Laval
- Department of Pediatrics I - Immunology and Pneumology/Cystic fibrosis, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Patrick Schlegel
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Christian Seitz
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbruecken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbruecken, Germany.,Department of Pharmacy, Saarland University, Saarbruecken, Germany
| | - Rupert Handgretinger
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany.,Department of Hematology, Oncology, Clinical Immunology, University of Tuebingen, Tuebingen, Germany
| | - Michael S D Kormann
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
11
|
Ehret F, Zhou CY, Alexander SC, Zhang D, Devaraj NK. Site-Specific Covalent Conjugation of Modified mRNA by tRNA Guanine Transglycosylase. Mol Pharm 2018; 15:737-742. [PMID: 28749687 DOI: 10.1021/acs.molpharmaceut.7b00356] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Modified mRNA (mod-mRNA) has recently been widely studied as the form of RNA useful for therapeutic applications due to its high stability and lowered immune response. Herein, we extend the scope of the recently established RNA-TAG (transglycosylation at guanosine) methodology, a novel approach for genetically encoded site-specific labeling of large mRNA transcripts, by employing mod-mRNA as substrate. As a proof of concept, we covalently attached a fluorescent probe to mCherry encoding mod-mRNA transcripts bearing 5-methylcytidine and/or pseudouridine substitutions with high labeling efficiencies. To provide a versatile labeling methodology with a wide range of possible applications, we employed a two-step strategy for functionalization of the mod-mRNA to highlight the therapeutic potential of this new methodology. We envision that this novel and facile labeling methodology of mod-RNA will have great potential in decorating both coding and noncoding therapeutic RNAs with a variety of diagnostic and functional moieties.
Collapse
Affiliation(s)
- Fabian Ehret
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - Cun Yu Zhou
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - Seth C Alexander
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - Dongyang Zhang
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| | - Neal K Devaraj
- Department of Chemistry and Biochemistry , University of California, San Diego , 9500 Gilman Drive , La Jolla , California 92093 , United States
| |
Collapse
|
12
|
Yoon JC. Evolving Mechanistic Views and Emerging Therapeutic Strategies for Cystic Fibrosis-Related Diabetes. J Endocr Soc 2017; 1:1386-1400. [PMID: 29264462 PMCID: PMC5686691 DOI: 10.1210/js.2017-00362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/25/2017] [Indexed: 12/19/2022] Open
Abstract
Diabetes is a common and important complication of cystic fibrosis, an autosomal recessive genetic disease due to mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Cystic fibrosis-related diabetes (CFRD) is associated with profound detrimental effects on the disease course and mortality and is expected to increase in prevalence as the survival of patients with cystic fibrosis continues to improve. Despite progress in the functional characterization of CFTR molecular defects, the mechanistic basis of CFRD is not well understood, in part because of the relative inaccessibility of the pancreatic tissue and the limited availability of representative animal models. This review presents a concise overview of the current understanding of CFRD pathogenesis and provides a cutting-edge update on novel findings from human and animal studies. Potential contributions from paracrine mechanisms and β-cell compensatory mechanisms are highlighted, as well as functional β-cell and α-cell defects, incretin defects, exocrine pancreatic insufficiency, and loss of islet cell mass. State-of-the-art and emerging treatment options are explored, including advances in insulin administration, CFTR modulators, cell replacement, gene replacement, and gene editing therapies.
Collapse
Affiliation(s)
- John C Yoon
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, California 95616
| |
Collapse
|
13
|
Kawasaki S, Fujita Y, Nagaike T, Tomita K, Saito H. Synthetic mRNA devices that detect endogenous proteins and distinguish mammalian cells. Nucleic Acids Res 2017; 45:e117. [PMID: 28525643 PMCID: PMC5499560 DOI: 10.1093/nar/gkx298] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 04/13/2017] [Indexed: 01/04/2023] Open
Abstract
Synthetic biology has great potential for future therapeutic applications including autonomous cell programming through the detection of protein signals and the production of desired outputs. Synthetic RNA devices are promising for this purpose. However, the number of available devices is limited due to the difficulty in the detection of endogenous proteins within a cell. Here, we show a strategy to construct synthetic mRNA devices that detect endogenous proteins in living cells, control translation and distinguish cell types. We engineered protein-binding aptamers that have increased stability in the secondary structures of their active conformation. The designed devices can efficiently respond to target proteins including human LIN28A and U1A proteins, while the original aptamers failed to do so. Moreover, mRNA delivery of an LIN28A-responsive device into human induced pluripotent stem cells (hiPSCs) revealed that we can distinguish living hiPSCs and differentiated cells by quantifying endogenous LIN28A protein expression level. Thus, our endogenous protein-driven RNA devices determine live-cell states and program mammalian cells based on intracellular protein information.
Collapse
Affiliation(s)
- Shunsuke Kawasaki
- Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.,Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshihiko Fujita
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takashi Nagaike
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Kozo Tomita
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
14
|
Tam YK, Madden TD, Hope MJ. Pieter Cullis’ quest for a lipid-based, fusogenic delivery system for nucleic acid therapeutics: success with siRNA so what about mRNA? J Drug Target 2016; 24:774-779. [DOI: 10.1080/1061186x.2016.1221955] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
15
|
Intracellular delivery of messenger RNA by recombinant PP7 virus-like particles carrying low molecular weight protamine. BMC Biotechnol 2016; 16:46. [PMID: 27233770 PMCID: PMC4884372 DOI: 10.1186/s12896-016-0274-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/11/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell-penetrating peptides (CPPs) have been widely used as carriers to transport different molecules into living cells, whereas messenger RNAs (mRNAs) have been utilized as target molecules for the prevention and treatment of various diseases. However, the instability of CPPs and mRNAs has limited their application. Bacteriophage PP7 virus-like particles (VLPs) may protect peptides and RNAs from degradation through displaying foreign peptides on their surface and encapsidating RNA linked with the pac site. RESULTS In this study, the cDNA of the PP7 coat protein single-chain dimer carrying low molecular weight protamine (LMWP) and the cDNA of green fluorescent protein (GFP) were inserted into two multiple cloning sites of pETDuet-1, respectively. PP7 VLPs carrying the LMWP peptide and GFP mRNA were subsequently expressed in Escherichia coli BL21 (DE3) with high yield and thermal stability, and were easily purified. The VLPs were also non-replicative, non-infectious, and non-toxic. Moreover, they penetrated the mouse prostate cancer cells RM-1 after 24 h incubation. Last, PP7 VLPs carrying the LMWP could encapsidate the GFP mRNA, which was translated into mature protein in mammalian cells. CONCLUSIONS Recombinant PP7 VLPs can be used simultaneously as a targeted delivery vector for both peptides and mRNA due to their abilities to package RNA and display peptides.
Collapse
|