1
|
Yang S, Zhang Q, Zhuang Y, Li J, Fu X. Plate centrifugation enhances the efficiency of polyethylenimine-based transfection and lentiviral infection. J Virol Methods 2024; 330:115039. [PMID: 39357745 DOI: 10.1016/j.jviromet.2024.115039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE To propose an efficient, reproducible, and consistent transgenic technology based on plate centrifugation, which is particularly useful for polyethylenimine (PEI) transfection and lentiviral infection. METHODS We optimized multiple factors that could contribute to transfection efficiency, such as the dosage of the PEI or DNA, the working solution buffer used for diluting the PEI or DNA, the incubation time for the PEI/DNA complexes, and the transfection time. RESULTS Plate centrifugation led to a 5.46-fold increase in the transfection efficiency of PEI-based transfection while maintaining the cell survival rate. Moreover, the average copy number of viral genes in each genome increased 4.96-fold with plate centrifugation. Plate centrifugation alters the spatial arrangement of the PEI/DNA complexes or lentiviruses, increasing the chances of these complexes or viruses coming into contact with target cells, ultimately resulting in improved transfection or infection efficiency. CONCLUSIONS We present a protocol based on plate centrifugation for transfection or lentiviral infection that is suitable for genetic modification of primary cells or stem cells.
Collapse
Affiliation(s)
- Shaozhe Yang
- Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China; Prenatal Diagnostic Center, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China
| | - Qingwei Zhang
- Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China; Prenatal Diagnostic Center, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China
| | - Yuan Zhuang
- Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China; Prenatal Diagnostic Center, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China
| | - Junfeng Li
- Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China; Prenatal Diagnostic Center, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China
| | - Xiuhong Fu
- Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China; Prenatal Diagnostic Center, Luohe Central Hospital, 54 Renmin East Road, Luohe 462000, PR China.
| |
Collapse
|
2
|
Setyawati DR, Azzahra K, Mardliyati E, Tarwadi, Maharani BY, Nurmeilis. Box-Behnken design assisted approach in optimizing lipid composition for cationic liposome formulation as gene carrier. Biochim Biophys Acta Gen Subj 2024; 1868:130705. [PMID: 39178921 DOI: 10.1016/j.bbagen.2024.130705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Cationic liposomes represent a promising non-viral carrier platform for gene delivery. The successful intracellular delivery of genes to the target cell is highly influenced by lipid compositions in the liposomal formulation. In the present study, a Box-Behnken design was applied to investigate the optimal lipid composition for the liposome-based transfection agent. METHODS The concentrations of DOTAP, DSPE-PEG, and cholesterol were set as independent factors. A total of 15 lipid compositions were generated and tested for specific responses, including particle size, encapsulation efficiency, cell viability, and cell transfection. The data were then analyzed to predict the optimal composition using response surface methodology (RSM). RESULTS The results for particle size, encapsulation efficiency, cell viability and fluorescence intensity ranged from 158.7 to 2064 nm, 48.19-95.72%, 81.50-122.67%, and 0.0-9.08, respectively. Compositions of liposome-based transfection agent without DOTAP, those without cholesterol, and those containing DSPE-PEG2000 with a molar ratio equal to or greater than that of cholesterol tended to exhibit low encapsulation efficiency. The ability of the liposome to complex DNA, as determined through electrophoresis gel retardation assay, showed that the composition without DOTAP produced DNA bands, indicating that the prepared liposomes had a less ability to complex DNA. The cytotoxicity test results indicated that all lipid compositions were considered non-toxic, as they exhibited >80% cell viability. The cell transfection assay demonstrated that the lipid composition containing a combination of DOTAP and cholesterol was able to transfect DNA into cells. According to response analysis, RSM predicted that the optimal lipid composition consisted of 2.75 μmol DOTAP and 0.91 μmol cholesterol, with a desirability value of 0.85. CONCLUSIONS Although the equation model is still acceptable for predicting the optimal lipid composition, further study is needed to obtain a model with higher desirability, such as by using more lipid compositions, increased replications, and different variable responses.
Collapse
Affiliation(s)
- Damai Ria Setyawati
- Research Center for Vaccine and Drugs, National Research and Innovation Agency, Jakarta, Indonesia
| | - Khairunnisa Azzahra
- Department of Pharmacy, Faculty of Health and Sciences, Universitas Islam Negeri Syarif Hidayatullah, Jakarta, Indonesia; Nano Center Indonesia, South Tangerang 15314, Indonesia
| | - Etik Mardliyati
- Research Center for Vaccine and Drugs, National Research and Innovation Agency, Jakarta, Indonesia.
| | - Tarwadi
- Research Center for Vaccine and Drugs, National Research and Innovation Agency, Jakarta, Indonesia
| | - Bismi Yasinta Maharani
- Research Center for Vaccine and Drugs, National Research and Innovation Agency, Jakarta, Indonesia
| | - Nurmeilis
- Department of Pharmacy, Faculty of Health and Sciences, Universitas Islam Negeri Syarif Hidayatullah, Jakarta, Indonesia
| |
Collapse
|
3
|
Gonzalez-Rivera JC, Galvan A, Ryder T, Milman M, Agarwal K, Kandari L, Khetan A. A high-titer scalable Chinese hamster ovary transient expression platform for production of biotherapeutics. Biotechnol Bioeng 2024; 121:3454-3470. [PMID: 39101569 DOI: 10.1002/bit.28817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024]
Abstract
Transient gene expression (TGE) in Chinese hamster ovary (CHO) cells offers a route to accelerate biologics development by delivering material weeks to months earlier than what is possible with conventional cell line development. However, low productivity, inconsistent product quality profiles, and scalability challenges have prevented its broader adoption. In this study, we develop a scalable CHO-based TGE system achieving 1.9 g/L of monoclonal antibody in an unmodified host. We integrated continuous flow-electroporation and alternate tangential flow (ATF) perfusion to enable an end-to-end closed system from N-1 perfusion to fed-batch 50-L bioreactor production. Optimization of both the ATF operation for three-in-one application-cell growth, buffer exchange, and cell mass concentration-and the flow-electroporation process, led to a platform for producing biotherapeutics using transiently transfected cells. We demonstrate scalability up to 50-L bioreactor, maintaining a titer over 1 g/L. We also show comparable quality between both transiently and stably produced material, and consistency across batches. The results confirm that purity, charge variants and N-glycan profiles are similar. Our study demonstrates the potential of CHO-based TGE platforms to accelerate biologics process development timelines and contributes evidence supporting its feasibility for manufacturing early clinical material, aiming to strengthen endorsement for TGE's wider implementation.
Collapse
Affiliation(s)
| | - Alberto Galvan
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Todd Ryder
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Monica Milman
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Kitty Agarwal
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Lakshmi Kandari
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Anurag Khetan
- Biologics Development, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| |
Collapse
|
4
|
Rocher EE, Luly KM, Tzeng SY, Sunshine JC, Green JJ. Efficient Polymeric Nanoparticle Gene Delivery Enabled Via Tri- and Tetrafunctional Branching. Biomacromolecules 2024. [PMID: 39466232 DOI: 10.1021/acs.biomac.4c00954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Poly(β-amino ester) (PBAE) nanoparticles (NPs) show great promise for nonviral gene delivery. Recent studies suggest branched PBAEs (BPBAEs) offer advantages over linear counterparts, but the effect of polymer structure has not been well investigated across many chemical constituents. Here, a library of BPBAEs was synthesized with tri- and tetrafunctional branching. These polymers self-assemble with DNA to form highly cationic, monodisperse NPs with notably small size (∼50 nm). Optimal transfection occurred with polymer structures that featured moderate PBAE branching, enabling complete DNA encapsulation, rapid NP uptake, and robust expression at low DNA doses and polymer amounts. Optimized NPs enabled efficient DNA delivery to diverse cell types in vitro while maintaining high cellular viability, demonstrating significant improvements over a well-performing linear PBAE counterpart. BPBAEs also facilitated efficient mRNA and siRNA delivery, highlighting the versatility of these structures and demonstrating the broad utility of BPBAE NPs as vectors for nucleic acid delivery.
Collapse
|
5
|
Chen X, Gao Y, Zhang Y. Allogeneic CAR-T cells for cancer immunotherapy. Immunotherapy 2024; 16:1079-1090. [PMID: 39378059 PMCID: PMC11492692 DOI: 10.1080/1750743x.2024.2408048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Autologous chimeric antigen receptor (CAR)-modified T (CAR-T) cell therapy has displayed high efficacy in the treatment of hematological malignancies. Up to now, 11 autologous CAR-T cell products have been approved for the management of malignancies globally. However, the application of autologous CAR-T cell therapy has many individual limitations, long time-consuming, highly cost, and the risk of manufacturing failure. Indeed, some patients would not benefit from autologous CAR-T cell products because of rapid disease progression. Allogeneic CAR-T cells especially universal CAR-T (U-CAR-T) cell therapy are superior to these challenges of autologous CAR-T cells. In this review, we describe basic study and clinical trials of U-CAR-T cell therapeutic methods for malignancies. In addition, we summarize the problems encountered and potential solutions.
Collapse
Affiliation(s)
- Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yaoxin Gao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, China
| |
Collapse
|
6
|
Hu Y, Tzeng SY, Cheng L, Lin J, Villabona-Rueda A, Yu S, Li S, Schneiderman Z, Zhu Y, Ma J, Wilson DR, Shannon SR, Warren T, Rui Y, Qiu C, Kavanagh EW, Luly KM, Zhang Y, Korinetz N, D’Alessio FR, Wang TH, Kokkoli E, Reddy SK, Luijten E, Green JJ, Mao HQ. Supramolecular assembly of polycation/mRNA nanoparticles and in vivo monocyte programming. Proc Natl Acad Sci U S A 2024; 121:e2400194121. [PMID: 39172792 PMCID: PMC11363337 DOI: 10.1073/pnas.2400194121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/19/2024] [Indexed: 08/24/2024] Open
Abstract
Size-dependent phagocytosis is a well-characterized phenomenon in monocytes and macrophages. However, this size effect for preferential gene delivery to these important cell targets has not been fully exploited because commonly adopted stabilization methods for electrostatically complexed nucleic acid nanoparticles, such as PEGylation and charge repulsion, typically arrest the vehicle size below 200 nm. Here, we bridge the technical gap in scalable synthesis of larger submicron gene delivery vehicles by electrostatic self-assembly of charged nanoparticles, facilitated by a polymer structurally designed to modulate internanoparticle Coulombic and van der Waals forces. Specifically, our strategy permits controlled assembly of small poly(β-amino ester)/messenger ribonucleic acid (mRNA) nanoparticles into particles with a size that is kinetically tunable between 200 and 1,000 nm with high colloidal stability in physiological media. We found that assembled particles with an average size of 400 nm safely and most efficiently transfect monocytes following intravenous administration and mediate their differentiation into macrophages in the periphery. When a CpG adjuvant is co-loaded into the particles with an antigen mRNA, the monocytes differentiate into inflammatory dendritic cells and prime adaptive anticancer immunity in the tumor-draining lymph node. This platform technology offers a unique ligand-independent, particle-size-mediated strategy for preferential mRNA delivery and enables therapeutic paradigms via monocyte programming.
Collapse
Affiliation(s)
- Yizong Hu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Leonardo Cheng
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Jinghan Lin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Andres Villabona-Rueda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Shuai Yu
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL60208
| | - Sixuan Li
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Zachary Schneiderman
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Yining Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Jingyao Ma
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - David R. Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Sydney R. Shannon
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Tiarra Warren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Yuan Rui
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Chenhu Qiu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Erin W. Kavanagh
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Kathryn M. Luly
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
| | - Yicheng Zhang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Nicole Korinetz
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Franco R. D’Alessio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Tza-Huei Wang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Efrosini Kokkoli
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Sashank K. Reddy
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Erik Luijten
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL60208
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL60208
- Department of Chemistry, Northwestern University, Evanston, IL60208
- Department of Physics and Astronomy, Northwestern University, Evanston, IL60208
| | - Jordan J. Green
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD21218
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD21231
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD21218
| |
Collapse
|
7
|
Dalal RJ, Oviedo F, Leyden MC, Reineke TM. Polymer design via SHAP and Bayesian machine learning optimizes pDNA and CRISPR ribonucleoprotein delivery. Chem Sci 2024; 15:7219-7228. [PMID: 38756796 PMCID: PMC11095369 DOI: 10.1039/d3sc06920f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/25/2024] [Indexed: 05/18/2024] Open
Abstract
We present the facile synthesis of a clickable polymer library with systematic variations in length, binary composition, pKa, and hydrophobicity (clog P) to optimize intracellular pDNA and CRISPR-Cas9 ribonucleoprotein (RNP) performance. We couple physicochemical characterization and machine learning to interpret quantitative structure-property relationships within the combinatorial design space. For the first time, we reveal unexpected disparate design parameters for nucleic acid carriers; via explainable machine learning on 432 formulations, we discover that lower polymer pKa and higher percentages of benzimidazole ethanethiol enhance pDNA delivery, yet polymer length and captamine cation identity improve RNP delivery. Closed-loop Bayesian optimization of 552 formulation ratios further enhances in vitro performance. The top three polymers yield a higher signal and stable transgene expression over 20 days in vivo, and a 1.7-fold enhancement over controls. Our facile coupling of synthesis, characterization, and machine analysis provides powerful tools to quantitate performance parameters accelerating next-generation vehicles for nucleic acid medicines.
Collapse
Affiliation(s)
- Rishad J Dalal
- Department of Chemistry, University of Minnesota Minneapolis Minnesota 55455 USA
| | | | - Michael C Leyden
- Department of Chemical Engineering and Materials Science, University of Minnesota Minneapolis Minnesota 55455 USA
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota Minneapolis Minnesota 55455 USA
| |
Collapse
|
8
|
Hu Y, Eder BA, Lin J, Li S, Zhu Y, Wang TH, Guo T, Mao HQ. Liter-scale manufacturing of shelf-stable plasmid DNA/PEI transfection particles for viral vector production. Mol Ther Methods Clin Dev 2024; 32:101194. [PMID: 38352269 PMCID: PMC10863326 DOI: 10.1016/j.omtm.2024.101194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
The transfection efficiency and stability of the delivery vehicles of plasmid DNA (pDNA) are critical metrics to ensure high-quality and high-yield production of viral vectors. We previously identified that the optimal size of pDNA/poly(ethylenimine) (PEI) transfection particles is 400-500 nm and developed a bottom-up assembly method to construct stable 400-nm pDNA/PEI particles and benchmarked their transfection efficiency in producing lentiviral vectors (LVVs). Here, we report scale-up production protocols for such transfection particles. Using a two-inlet confined impinging jet (CIJ) mixer with a dual syringe pump set-up, we produced a 1-L batch at a flow rate of 100 mL/min, and further scaled up this process with a larger CIJ mixer and a dual peristaltic pump array, allowing for continuous production at a flow rate of 1 L/min without a lot size limit. We demonstrated the scalability of this process with a 5-L lot and validated the quality of these 400-nm transfection particles against the target product profile, including physical properties, shelf and on-bench stability, transfection efficiency, and LVV production yield in both 15-mL bench culture and 2-L bioreactor runs. These results confirm the potential of this particle assembly process as a scalable manufacturing platform for viral vector production.
Collapse
Affiliation(s)
- Yizong Hu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | - Jinghan Lin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sixuan Li
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yining Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Tza-Huei Wang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ting Guo
- 2seventy bio, Inc., Cambridge, MA 02142, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
9
|
Kreofsky NW, Roy P, Brown ME, Perez U, Leighton RE, Frontiera RR, Reineke TM. Cinchona Alkaloid Polymers Demonstrate Highly Efficient Gene Delivery Dependent on Stereochemistry, Methoxy Substitution, and Length. Biomacromolecules 2024; 25:486-501. [PMID: 38150323 DOI: 10.1021/acs.biomac.3c01099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Nucleic acid delivery with cationic polymers is a promising alternative to expensive viral-based methods; however, it often suffers from a lower performance. Herein, we present a highly efficient delivery system based on cinchona alkaloid natural products copolymerized with 2-hydroxyethyl acrylate. Cinchona alkaloids are an attractive monomer class for gene delivery applications, given their ability to bind to DNA via both electrostatics and intercalation. To uncover the structure-activity profile of the system, four structurally similar cinchona alkaloids were incorporated into polymers: quinine, quinidine, cinchonine, and cinchonidine. These polymers differed in the chain length, the presence or absence of a pendant methoxy group, and stereochemistry, all of which were found to alter gene delivery performance and the ways in which the polymers overcome biological barriers to transfection. Longer polymers that contained the methoxy-bearing cinchona alkaloids (i.e., quinine and quinidine) were found to have the best performance. These polymers exhibited the tightest DNA binding, largest and most abundant DNA-polymer complexes, and best endosomal escape thanks to their increased buffering capacity and closest nuclear proximity of the payload. Overall, this work highlights the remarkable efficiency of polymer systems that incorporate cinchona alkaloid natural products while demonstrating the profound impact that small structural changes can have on overcoming biological hurdles associated with gene delivery.
Collapse
Affiliation(s)
- Nicholas W Kreofsky
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Punarbasu Roy
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mary E Brown
- University Imaging Centers, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ulises Perez
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ryan E Leighton
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Renee R Frontiera
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
10
|
Jerca FA, Muntean C, Remaut K, Jerca VV, Raemdonck K, Hoogenboom R. Cationic amino-acid functionalized polymethacrylamide vectors for siRNA transfection based on modification of poly(2-isopropenyl-2-oxazoline). J Control Release 2023; 364:687-699. [PMID: 37935258 DOI: 10.1016/j.jconrel.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023]
Abstract
Poly(2-isopropenyl-2-oxazoline) (PiPOx) is a functional polymer showing great potential for the development of smart biomaterials. The straightforward synthesis and post-polymerization functionalization of PiPOx offers many opportunities for tailoring the properties of the polymer towards biomaterials. In this study we report for the first time PiPOx-based cationic charged polymethacrylamides with amino acid side chains that can complex siRNA and promote transfection in vitro. Therefore, PiPOx was fully modified via ring opening addition reactions with the carboxylic acid groups of a series of N-Boc-L-amino acids and their reaction kinetics were investigated. Based on the determined kinetic constants, another series of PiPOx-based copolymers with balanced hydrophilic/hydrophobic content of N-Boc-L-amino acids were obtained via one-pot modification reaction with two different N-Boc-L-amino acids. The N-Boc protected homopolymers and related copolymers were deprotected to obtain (co)polymers with the targeted side chain cationic charged units. The (co)polymers' structures were fully investigated via FT-IR and 1H NMR spectroscopy, size exclusion chromatography (SEC), and TGA-DSC-MS analysis. The polarimetry measurements revealed that the homopolymers retain their chiroptical properties after post-modification, and a sign inversion is noticed from (L) N-Boc-protected analogues to (D) for the TFA cationic charged homopolymers. Generally, cationically charged homopolymers with hydrophilic amino acids on the side chain showed efficient complexation of siRNA, but poor transfection while cationic copolymers having both tryptophan and valine or proline side chains revealed moderate siRNA binding, high transfection efficiency (> 90% of the cells) and potent gene silencing with IC50 values down to 5.5 nM. Particularly, these cationic copolymers showed higher gene silencing potency as compared to the commercial JetPRIME® reference, without reducing cell viability in the concentration range used for transfection, making this a very interesting system for in vitro siRNA transfection.
Collapse
Affiliation(s)
- Florica Adriana Jerca
- Smart Organic Materials Group, "Costin D. Nenitzescu" Institute of Organic and Supramolecular Chemistry, Romanian Academy, 202B Spl. Independentei CP 35-108, 060023 Bucharest, Romania; Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium.
| | - Cristina Muntean
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Valentin Victor Jerca
- Smart Organic Materials Group, "Costin D. Nenitzescu" Institute of Organic and Supramolecular Chemistry, Romanian Academy, 202B Spl. Independentei CP 35-108, 060023 Bucharest, Romania; Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, B-9000 Ghent, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, B-9000 Ghent, Belgium.
| |
Collapse
|
11
|
Sinani G, Durgun ME, Cevher E, Özsoy Y. Polymeric-Micelle-Based Delivery Systems for Nucleic Acids. Pharmaceutics 2023; 15:2021. [PMID: 37631235 PMCID: PMC10457940 DOI: 10.3390/pharmaceutics15082021] [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: 05/13/2023] [Revised: 07/11/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Nucleic acids can modulate gene expression specifically. They are increasingly being utilized and show huge potential for the prevention or treatment of various diseases. However, the clinical translation of nucleic acids faces many challenges due to their rapid clearance after administration, low stability in physiological fluids and limited cellular uptake, which is associated with an inability to reach the intracellular target site and poor efficacy. For many years, tremendous efforts have been made to design appropriate delivery systems that enable the safe and effective delivery of nucleic acids at the target site to achieve high therapeutic outcomes. Among the different delivery platforms investigated, polymeric micelles have emerged as suitable delivery vehicles due to the versatility of their structures and the possibility to tailor their composition for overcoming extracellular and intracellular barriers, thus enhancing therapeutic efficacy. Many strategies, such as the addition of stimuli-sensitive groups or specific ligands, can be used to facilitate the delivery of various nucleic acids and improve targeting and accumulation at the site of action while protecting nucleic acids from degradation and promoting their cellular uptake. Furthermore, polymeric micelles can be used to deliver both chemotherapeutic drugs and nucleic acid therapeutics simultaneously to achieve synergistic combination treatment. This review focuses on the design approaches and current developments in polymeric micelles for the delivery of nucleic acids. The different preparation methods and characteristic features of polymeric micelles are covered. The current state of the art of polymeric micelles as carriers for nucleic acids is discussed while highlighting the delivery challenges of nucleic acids and how to overcome them and how to improve the safety and efficacy of nucleic acids after local or systemic administration.
Collapse
Affiliation(s)
- Genada Sinani
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Altinbas University, 34147 Istanbul, Türkiye;
| | - Meltem Ezgi Durgun
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| | - Erdal Cevher
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| | - Yıldız Özsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, 34126 Istanbul, Türkiye; (M.E.D.); (E.C.)
| |
Collapse
|
12
|
Agbaria M, Jbara-Agbaria D, Grad E, Ben-David-Naim M, Aizik G, Golomb G. Nanoparticles of VAV1 siRNA combined with LL37 peptide for the treatment of pancreatic cancer. J Control Release 2023; 355:312-326. [PMID: 36736910 DOI: 10.1016/j.jconrel.2023.01.084] [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: 11/16/2022] [Revised: 01/10/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the leading causes of cancer-related death, and it is highly resistant to therapy owing to its unique extracellular matrix. VAV1 protein, overexpressed in several cancer diseases including pancreatic cancer (PC), increases tumor proliferation and enhances metastases formation, which are associated with decreased survival. We hypothesized that an additive anti-tumor effect could be obtained by co-encapsulating in PLGA nanoparticles (NPs), the negatively charged siRNA against VAV1 (siVAV1) with the positively charged anti-tumor LL37 peptide, as a counter-ion. Several types of NPs were formulated and were characterized for their physicochemical properties, cellular internalization, and bioactivity in vitro. NPs' biodistribution, toxicity, and bioactivity were examined in a mice PDAC model. An optimal siVAV1 formulation (siVAV1-LL37 NPs) was characterized with desirable physicochemical properties in terms of nano-size, low polydispersity index (PDI), neutral surface charge, high siVAV1 encapsulation efficiency, spherical shape, and long-term shelf-life stability. Cell assays demonstrated rapid engulfment by PC cells, a specific and significant dose-dependent proliferation inhibition, as well as knockdown of VAV1 mRNA levels and migration inhibition in VAV1+ cells. Treatment with siVAV1-LL37 NPs in the mice PDAC model revealed marked accumulation of NPs in the liver and in the tumor, resulting in an increased survival rate following suppression of tumor growth and metastases, mediated via the knockdown of both VAV1 mRNA and protein levels. This proof-of-concept study validates our hypothesis of an additive effect in the treatment of PC facilitated by co-encapsulating siVAV1 in NPs with LL37 serving a dual role as a counter ion as well as an anti-tumor agent.
Collapse
Affiliation(s)
- Majd Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Doaa Jbara-Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Etty Grad
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Meital Ben-David-Naim
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Gil Aizik
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| |
Collapse
|
13
|
Casper J, Nicolle L, Willimann M, Kuzucu EÜ, Tran A, Robin P, Detampel P, Grisch-Chan HM, Thöny B, Huwyler J, Gerber-Lemaire S. Core-Shell Structured Chitosan-Polyethylenimine Nanoparticles for Gene Delivery: Improved Stability, Cellular Uptake, and Transfection Efficiency. Macromol Biosci 2023; 23:e2200314. [PMID: 36200651 DOI: 10.1002/mabi.202200314] [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: 07/28/2022] [Revised: 09/26/2022] [Indexed: 01/19/2023]
Abstract
The delivery of nucleic acids relies on vectors that condense and encapsulate their cargo. Especially nonviral gene delivery systems are of increasing interest. However, low transgene expression levels and limited tolerability of these systems remain a challenge. The improvement of nucleic acid delivery using depolymerized chitosan-polyethylenimine DNA complexes (dCS-PEI/DNA) is investigated. The secore complexes are further combined with chitosan-based shells and functionalized with polyethylene glycol (PEG) and cell penetrating peptides. This modular approach allows to evaluate the effect of functional shell components on physicochemical particle characteristics and biological effects. The optimized ternary complex combines a core-dCS-linear PEI/DNA complex with a shell consisting of dCS-PEG-COOH, which results in improved nucleic acid encapsulation, cellular uptake and transfection potency in human hepatoma HuH-7cells and murine primary hepatocytes. Effects on transgene expression are confirmed in wild-type mice following retrograde intrabiliary infusion. After administration of only 100 ng complexed DNA, ternary complexes induced a high reporter gene signal for three days. It is concluded that ternary coreshell structured nanoparticles comprising functionalized chitosan can be used for in vitro andin vivo gene delivery.
Collapse
Affiliation(s)
- Jens Casper
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, CH-4056, Switzerland
| | - Laura Nicolle
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
| | - Melanie Willimann
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, Zürich, CH-8032, Switzerland
| | - Evrim Ümit Kuzucu
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, CH-4056, Switzerland
| | - Alan Tran
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, CH-4056, Switzerland
| | - Perrine Robin
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
| | - Pascal Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, CH-4056, Switzerland
| | - Hiu Man Grisch-Chan
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, Zürich, CH-8032, Switzerland
| | - Beat Thöny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, Zürich, CH-8032, Switzerland
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, CH-4056, Switzerland
| | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, CH-1015, Switzerland
| |
Collapse
|
14
|
Protopapa G, Bono N, Visone R, D'Alessandro F, Rasponi M, Candiani G. A new microfluidic platform for the highly reproducible preparation of non-viral gene delivery complexes. LAB ON A CHIP 2022; 23:136-145. [PMID: 36477137 DOI: 10.1039/d2lc00744d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Transfection describes the delivery of exogenous nucleic acids (NAs) to cells utilizing non-viral means. In the last few decades, scientists have been doing their utmost to design ever more effective transfection reagents. These are eventually mixed with NAs to give rise to gene delivery complexes, which must undergo characterization, testing, and further refinement through the sequential reiteration of these steps. Unfortunately, although microfluidics offers distinct advantages over the canonical approaches to preparing particles, the systems available do not address the most frequent and practical quest for the simultaneous generation of multiple polymer-to-NA ratios (N/Ps). Herein, we developed a user-friendly microfluidic cartridge to repeatably prepare non-viral gene delivery particles and screen across a range of seven N/Ps at once or significant volumes of polyplexes at a given N/P. The microchip is equipped with a chaotic serial dilution generator for the automatic linear dilution of the polymer to the downstream area, which encompasses the NA divider to dispense equal amounts of DNA to the mixing area, enabling the formation of particles at seven N/Ps eventually collected in individual built-in tanks. This is the first example of a stand-alone microfluidic cartridge for the fast and repeatable preparation of non-viral gene delivery complexes at different N/Ps and their storage.
Collapse
Affiliation(s)
- Giovanni Protopapa
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy.
| | - Nina Bono
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy.
| | - Roberta Visone
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Fabio D'Alessandro
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy.
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Gabriele Candiani
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy.
| |
Collapse
|
15
|
Dalal RJ, Ohnsorg ML, Panda S, Reineke TM. Hydrophilic Surface Modification of Cationic Unimolecular Bottlebrush Vectors Moderate pDNA and RNP Bottleplex Stability and Delivery Efficacy. Biomacromolecules 2022; 23:5179-5192. [PMID: 36445696 DOI: 10.1021/acs.biomac.2c00999] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A cationic unimolecular bottlebrush polymer with chemically modified end-groups was synthesized to understand the impact of hydrophilicity on colloidal stability, nucleic acid delivery performance, and toxicity. The bottlebrush polymer template was synthesized using grafting-through techniques and was therefore composed of a polynorbornene backbone with poly(2-(dimethylamino)ethyl methacrylate) side chains with dodecyl trithiocarbonate end-groups. Postpolymerization modification was performed to fully remove the end-groups or install hydroxy and methoxy poly(ethylene glycol) functional groups on the bottlebrush exterior. The bottlebrush family was preformulated with biological payloads of pDNA and CRISPR-Cas9 RNP in both water and PBS to understand binding, aggregation kinetics, cytotoxicity, and delivery efficacy. Increasing end-group hydrophilicity and preformulation of bottleplexes in PBS increased colloidal stability and cellular viability; however, this did not always result in increased transfection efficiency. The bottlebrush family exemplifies how formulation conditions, polymer loading, and end-group functionality of bottlebrushes can be tuned to balance expression with cytotoxicity ratios and result in enhanced overall performance.
Collapse
Affiliation(s)
- Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Monica L Ohnsorg
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Sidharth Panda
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
16
|
Novel Non-Viral Vectors Based on Pluronic ® F68PEI with Application in Oncology Field. Polymers (Basel) 2022; 14:polym14235315. [PMID: 36501709 PMCID: PMC9739301 DOI: 10.3390/polym14235315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/07/2022] Open
Abstract
Copolymers composed of low-molecular-weight polyethylenimine (PEI) and amphiphilic Pluronics® are safe and efficient non-viral vectors for pDNA transfection. A variety of Pluronic® properties provides a base for tailoring transfection efficacy in combination with the unique biological activity of this polymer group. In this study, we describe the preparation of new copolymers based on hydrophilic Pluronic® F68 and PEI (F68PEI). F68PEI polyplexes obtained by doping with free F68 (1:2 and 1:5 w/w) allowed for fine-tuning of physicochemical properties and transfection activity, demonstrating improved in vitro transfection of the human bone osteosarcoma epithelial (U2OS) and oral squamous cell carcinoma (SCC-9) cells when compared to the parent formulation, F68PEI. Although all tested systems condensed pDNA at varying polymer/DNA charge ratios (N/P, 5/1−100/1), the addition of free F68 (1:5 w/w) resulted in the formation of smaller polyplexes (<200 nm). Analysis of polyplex properties by transmission electron microscopy and dynamic light scattering revealed varied polyplex morphology. Transfection potential was also found to be cell-dependent and significantly higher in SCC-9 cells compared to the control bPEI25k cells, as especially evident at higher N/P ratios (>25). The observed selectivity towards transfection of SSC-9 cells might represent a base for further optimization of a cell-specific transfection vehicle.
Collapse
|
17
|
Aranda-Barradas ME, Trejo-López SE, Real AD, Álvarez-Almazán S, Méndez-Albores A, García-Tovar CG, González-Díaz FR, Miranda-Castro SP. Effect of molecular weight of chitosan on the physicochemical, morphological, and biological properties of polyplex nanoparticles intended for gene delivery. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2022. [DOI: 10.1016/j.carpta.2022.100228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
18
|
Development and evaluation of polyethylenimine polyplexes as non-viral vectors for delivery of plasmid DNA encoding shRNA against STAT3 activity into triple negative breast cancer cells. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
19
|
Celebi Torabfam G, Yetisgin AA, Erdem C, Cayli A, Kutlu O, Cetinel S. A feasibility study of different commercially available serum-free mediums to enhance lentivirus and adeno-associated virus production in HEK 293 suspension cells. Cytotechnology 2022; 74:635-655. [PMID: 36389283 PMCID: PMC9652196 DOI: 10.1007/s10616-022-00551-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/30/2022] [Indexed: 02/02/2023] Open
Abstract
Lentivirus and adeno-associated viruses are invaluable tools for biotechnology applications due to their genetic material delivery abilities both in vitro and in vivo. However, their large-scale productions with Good Manufacturing Practices yield low efficiency when adherent and serum dependent HEK293 (Human Embryonic Kidney) cells are used as the host. To increase production efficiency, HEK293 cells are adapted to grow in suspension using commercially available and chemically defined serum-free mediums. Suspended cells can be transiently transfected for viral vector production; however, significant improvements are still needed to increase yield and thereby cost effectiveness. Here, we evaluated four most preferred commercially available mediums that are IVY, FreeStyle293, LV-MAX, and BalanCD HEK293 for the transient transfection feasibility of lentiviral (LV) and adeno-associated virus serotype 2 (AAV2) production in FlorabioHEK293 suspension cells. The highest transfection efficiency was over 90% and obtained by using polyethyleneimine (PEI) 25 K and by media adaptation in IVY without using any transfection enhancer. For the first time the feasibility of HEK293 cells, which were adapted to grow in suspension culture by Florabio and IVY media, were tested for virus production. This study demonstrates the best transfection medium for scalable and optimized production of Lentivirus and Adeno-Associated Virus in suspended HEK293 cell culture. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-022-00551-1.
Collapse
Affiliation(s)
- Gizem Celebi Torabfam
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, 34956 Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, Istanbul, 34956 Turkey
| | - Abuzer Alp Yetisgin
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, 34956 Turkey
- Faculty of Engineering and Natural Sciences, Materials Science and Nano Engineering, Sabanci University, Istanbul, 34956 Turkey
| | - Cem Erdem
- FloraBio Technology, Urla, 35430 İzmir Turkey
| | - Aziz Cayli
- FloraBio Technology, Urla, 35430 İzmir Turkey
| | - Ozlem Kutlu
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, 34956 Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, Istanbul, 34956 Turkey
| | - Sibel Cetinel
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, 34956 Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, Istanbul, 34956 Turkey
| |
Collapse
|
20
|
Luly KM, Yang H, Lee SJ, Wang W, Ludwig SD, Tarbox HE, Wilson DR, Green JJ, Spangler JB. Poly(Beta-Amino Ester)s as High-Yield Transfection Reagents for Recombinant Protein Production. Int J Nanomedicine 2022; 17:4469-4479. [PMID: 36176585 PMCID: PMC9514136 DOI: 10.2147/ijn.s377371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Transient transfection is an essential tool for recombinant protein production, as it allows rapid screening for expression without stable integration of genetic material into a target cell genome. Poly(ethylenimine) (PEI) is the current gold standard for transient gene transfer, but transfection efficiency and the resulting protein yield are limited by the polymer’s toxicity. This study investigated the use of a class of cationic polymers, poly(beta-amino ester)s (PBAEs), as reagents for transient transfection in comparison to linear 25 kDa PEI, a commonly used transfection reagent. Methods Transfection efficiency and protein production were assessed in human embryonic kidney 293F (HEK) and Chinese hamster ovary-S (CHO) cell suspensions using PBAE-based nanoparticles in comparison to linear 25 kDa PEI. Production of both a cytosolic reporter and secreted antibodies was investigated. Results In both HEK and CHO cells, several PBAEs demonstrated superior transfection efficiency and enhanced production of a cytosolic reporter compared to linear 25 kDa PEI. This result extended to secreted proteins, as a model PBAE increased the production of 3 different secreted antibodies compared to linear 25 kDa PEI at culture scales ranging from 20 to 2000 mL. In particular, non-viral gene transfer using the lead PBAE/plasmid DNA nanoparticle formulation led to robust transfection of mammalian cells across different constructs, doses, volumes, and cell types. Conclusion These results show that PBAEs enhance transfection efficiency and increase protein yield compared to a widespread commercially available reagent, making them attractive candidates as reagents for use in recombinant protein production.
Collapse
Affiliation(s)
- Kathryn M Luly
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huilin Yang
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen J Lee
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Wentao Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth D Ludwig
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Haley E Tarbox
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - David R Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Neurosurgery and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Materials Science & Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jamie B Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Sun W, Jiang Z, Jiang W, Yang R. Universal chimeric antigen receptor T cell therapy - The future of cell therapy: A review providing clinical evidence. Cancer Treat Res Commun 2022; 33:100638. [PMID: 36184307 DOI: 10.1016/j.ctarc.2022.100638] [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: 07/29/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 12/14/2022]
Abstract
Autologous CAR-T therapy has shown promising outcomes in the treatment of tumors, particularly hematological malignancies over the past years. However, the application of autologous CAR-T therapy is limited, due to undesirable patient and/or peripheral blood characteristics, the high cost and long time period of manufacturing, and other challenges. Universal CAR-T therapy could overcome major limitations of autologous CAR-T therapy. In this review, we described the research and development status of universal CAR-T therapy for hematological malignancies. In addition, we also summarized the challenges had been encountered and the current solutions.
Collapse
Affiliation(s)
| | | | - Wen Jiang
- Gobroad Healthcare Group, Beijing, China
| | - Rui Yang
- Gobroad Healthcare Group, Beijing, China.
| |
Collapse
|
22
|
Agafonova L, Zhdanov D, Gladilina Y, Kanashenko S, Shumyantseva V. A pilot study on an electrochemical approach for assessing transient DNA transfection in eukaryotic cells. J Electroanal Chem (Lausanne) 2022. [DOI: 10.1016/j.jelechem.2022.116635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
23
|
Shayestehfar M, Farahi S, Kheiri Yeganeh Azar B, Memari A, Baluchnejadmojarad T, Faghihi F. Generating Human Induced Pluripotent Stem Cell Via Low-Dose Polyethylenimine-Mediated Transfection: An Optimized Protocol. DNA Cell Biol 2022; 41:903-916. [PMID: 35984994 DOI: 10.1089/dna.2022.0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human dermal fibroblasts (HDFs) can be reprogrammed through different strategies to generate human induced pluripotent stem cells (hiPSCs). However, most of these strategies require high-cost materials and specific equipment not readily accessible in most laboratories. Hence, liposomal and virus-based techniques can replace with polyethylenimine (PEI)-mediated transfection to overcome these challenges. However, few researchers have addressed the PEI's ability to transfect HDFs. This study used PEI reagent to transfer oriP/EBNA1-based vector into HDFs to produce hiPSC lines. We first described conditions allowing the efficient transfection of HDFs with low cytotoxicity and without specific types of equipment and optimized several parameters relevant to the transfection procedure. We then monitored the effect of different N/P ratios on transfection efficiency and cytotoxicity using flow cytometry and fluorescent microscopy. By the results, we found that transfection efficiency was greatly affected by plasmid DNA concentration, PEI concentration, order of combining reagents, serum presence in polyplexes, and the duration of serum starvations. Moreover, using the optimized condition, we found that the N/P ratio of 3 achieved the highest percentage of HDFs positive for green fluorescent protein plasmid (∼40%) with minimal cell toxicity. We finally generated hiPSCs using the optimized protocol and oriP/EBNA1-based vectors. We confirmed hiPSC formation by characterizing tests: alkaline phosphatase staining, immunocytochemistry assay, real-time PCR analysis, in vitro differentiation into three germ layers, and karyotyping test. In conclusion, our results indicated that 25 kDa branched PEI could efficiently transfect HDFs toward generating hiPSCs via a simple, cost-effective, and optimized condition.
Collapse
Affiliation(s)
- Monir Shayestehfar
- Department of Neuroscience, Faculty of Advanced Technology in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Farahi
- Biotechnology Department, Shahid Beheshti University of medical science, Tehran, Iran
| | - Behjat Kheiri Yeganeh Azar
- Department of Molecular Medicine, Faculty of Advanced Technology in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Memari
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Ponti F, Bono N, Russo L, Bigini P, Mantovani D, Candiani G. Vibropolyfection: coupling polymer-mediated gene delivery to mechanical stimulation to enhance transfection of adherent cells. J Nanobiotechnology 2022; 20:363. [PMID: 35933375 PMCID: PMC9356458 DOI: 10.1186/s12951-022-01571-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/22/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND With the success of recent non-viral gene delivery-based COVID-19 vaccines, nanovectors have gained some public acceptance and come to the forefront of advanced therapies. Unfortunately, the relatively low ability of the vectors to overcome cellular barriers adversely affects their effectiveness. Scientists have thus been striving to develop ever more effective gene delivery vectors, but the results are still far from satisfactory. Therefore, developing novel strategies is probably the only way forward to bring about genuine change. Herein, we devise a brand-new gene delivery strategy to boost dramatically the transfection efficiency of two gold standard nucleic acid (NA)/polymer nanoparticles (polyplexes) in vitro. RESULTS We conceived a device to generate milli-to-nanoscale vibrational cues as a function of the frequency set, and deliver vertical uniaxial displacements to adherent cells in culture. A short-lived high-frequency vibrational load (t = 5 min, f = 1,000 Hz) caused abrupt and extensive plasmalemma outgrowths but was safe for cells as neither cell proliferation rate nor viability was affected. Cells took about 1 hr to revert to quasi-naïve morphology through plasma membrane remodeling. In turn, this eventually triggered the mechano-activated clathrin-mediated endocytic pathway and made cells more apt to internalize polyplexes, resulting in transfection efficiencies increased from 10-to-100-fold. Noteworthy, these results were obtained transfecting three cell lines and hard-to-transfect primary cells. CONCLUSIONS In this work, we focus on a new technology to enhance the intracellular delivery of NAs and improve the transfection efficiency of non-viral vectors through priming adherent cells with a short vibrational stimulation. This study paves the way for capitalizing on physical cell stimulation(s) to significantly raise the effectiveness of gene delivery vectors in vitro and ex vivo.
Collapse
Affiliation(s)
- Federica Ponti
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
- Laboratory for Biomaterials and Bioengineering, CRC Tier I, Department of Min-Met-Mat Engineering and CHU de Québec Research Center, Division of Regenerative Medicine, Laval University, Quebec, QC, Canada
| | - Nina Bono
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy
| | - Luca Russo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milan, Italy
| | - Paolo Bigini
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milan, Italy
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, CRC Tier I, Department of Min-Met-Mat Engineering and CHU de Québec Research Center, Division of Regenerative Medicine, Laval University, Quebec, QC, Canada
| | - Gabriele Candiani
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Milan, Italy.
| |
Collapse
|
25
|
Mirón-Barroso S, Correia JS, Frampton AE, Lythgoe MP, Clark J, Tookman L, Ottaviani S, Castellano L, Porter AE, Georgiou TK, Krell J. Polymeric Carriers for Delivery of RNA Cancer Therapeutics. Noncoding RNA 2022; 8:ncrna8040058. [PMID: 36005826 PMCID: PMC9412371 DOI: 10.3390/ncrna8040058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/15/2022] [Accepted: 07/25/2022] [Indexed: 12/04/2022] Open
Abstract
As research uncovers the underpinnings of cancer biology, new targeted therapies have been developed. Many of these therapies are small molecules, such as kinase inhibitors, that target specific proteins; however, only 1% of the genome encodes for proteins and only a subset of these proteins has ‘druggable’ active binding sites. In recent decades, RNA therapeutics have gained popularity due to their ability to affect targets that small molecules cannot. Additionally, they can be manufactured more rapidly and cost-effectively than small molecules or recombinant proteins. RNA therapeutics can be synthesised chemically and altered quickly, which can enable a more personalised approach to cancer treatment. Even though a wide range of RNA therapeutics are being developed for various indications in the oncology setting, none has reached the clinic to date. One of the main reasons for this is attributed to the lack of safe and effective delivery systems for this type of therapeutic. This review focuses on current strategies to overcome these challenges and enable the clinical utility of these novel therapeutic agents in the cancer clinic.
Collapse
Affiliation(s)
- Sofía Mirón-Barroso
- Department of Surgery and Cancer, Imperial College, London W12 0HS, UK; (A.E.F.); (M.P.L.); (J.C.); (L.T.); (J.K.)
- Correspondence:
| | - Joana S. Correia
- Department of Materials, Imperial College London, London SW7 2AZ, UK; (J.S.C.); (A.E.P.); (T.K.G.)
| | - Adam E. Frampton
- Department of Surgery and Cancer, Imperial College, London W12 0HS, UK; (A.E.F.); (M.P.L.); (J.C.); (L.T.); (J.K.)
- Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK
| | - Mark P. Lythgoe
- Department of Surgery and Cancer, Imperial College, London W12 0HS, UK; (A.E.F.); (M.P.L.); (J.C.); (L.T.); (J.K.)
| | - James Clark
- Department of Surgery and Cancer, Imperial College, London W12 0HS, UK; (A.E.F.); (M.P.L.); (J.C.); (L.T.); (J.K.)
| | - Laura Tookman
- Department of Surgery and Cancer, Imperial College, London W12 0HS, UK; (A.E.F.); (M.P.L.); (J.C.); (L.T.); (J.K.)
| | - Silvia Ottaviani
- Department of Biosciences, Nottingham Trent University, Nottingham NG1 4FQ, UK;
| | | | - Alexandra E. Porter
- Department of Materials, Imperial College London, London SW7 2AZ, UK; (J.S.C.); (A.E.P.); (T.K.G.)
| | - Theoni K. Georgiou
- Department of Materials, Imperial College London, London SW7 2AZ, UK; (J.S.C.); (A.E.P.); (T.K.G.)
| | - Jonathan Krell
- Department of Surgery and Cancer, Imperial College, London W12 0HS, UK; (A.E.F.); (M.P.L.); (J.C.); (L.T.); (J.K.)
| |
Collapse
|
26
|
Dirksmeyer T, Stahl P, Vallet C, Knauer S, Giese M, Schmuck C, Hirschhäuser C. Advances towards Cell‐Specific Gene Transfection: A Small‐Molecule Approach Allows Order‐of‐Magnitude Selectivity. Chemistry 2022; 28:e202104618. [PMID: 35604769 PMCID: PMC9401007 DOI: 10.1002/chem.202104618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Indexed: 11/09/2022]
Abstract
A transfection vector that can home in on tumors is reported. Whereas previous vectors that allow moderately cell selective gene transfection used larger systems, this small‐molecule approach paved the way for precise structure‐activity relationship optimization. For this, biotin, which mediates cell selectivity, was combined with the potent DNA‐binding motif tetralysine‐guanidinocarbonypyrrol via a hydrophilic linker, thus enabling SAR‐based optimization. The new vector mediated biotin receptor (BR)‐selective transfection of cell lines with different BR expression levels. Computer‐based analyses of microscopy images revealed a preference of one order of magnitude for the BR‐positive cell lines over the BR‐negative controls.
Collapse
Affiliation(s)
- Thies Dirksmeyer
- Institute of Organic Chemistry University of Duisburg-Essen 45117 Essen Germany
| | - Paul Stahl
- Institute of Biology University of Duisburg-Essen 45117 Essen Germany
| | - Cecilia Vallet
- Institute of Biology University of Duisburg-Essen 45117 Essen Germany
| | - Shirley Knauer
- Institute of Biology University of Duisburg-Essen 45117 Essen Germany
| | - Michael Giese
- Institute of Organic Chemistry University of Duisburg-Essen 45117 Essen Germany
| | - Carsten Schmuck
- Institute of Organic Chemistry University of Duisburg-Essen 45117 Essen Germany
| | | |
Collapse
|
27
|
Evans CW, Ho D, Marlow JB, King JJ, Hee C, Wong LN, Atkin R, Smith NM, Warr GG, Norret M, Iyer KS. Intracellular Communication between Synthetic Macromolecules. J Am Chem Soc 2022; 144:14112-14120. [PMID: 35901278 DOI: 10.1021/jacs.2c02793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Non-viral delivery is an important strategy for selective and efficient gene therapy, immunization, and RNA interference, which overcomes problems of genotoxicity and inherent immunogenicity associated with viral vectors. Liposomes and polymers are compelling candidates as carriers for intracellular, non-viral delivery, but maximal efficiencies of around 1% have been reported for the most advanced non-viral carriers. Here, we develop a library of dendronized bottlebrush polymers with controlled defects, displaying a level of precision surpassed only by biological molecules like DNA, RNA, and proteins. We test concurrent and competitive delivery of DNA and show for the first time that, while intracellular communication is thought to be an exclusively biomolecular phenomenon, such communication between synthetic macromolecular complexes can also take place. Our findings challenge the assumption that delivery agents behave as bystanders that enable transfection by passive intracellular release of genetic cargo and improve upon coarse strategies in intracellular carrier design lacking control over polymer sequence, architecture, and composition, leading to a hit-or-miss outcome. Understanding the communication that takes place between macromolecules will help improve the design of non-viral delivery agents and facilitate translation of genome engineering, vaccines, and nucleic acid-based therapies.
Collapse
Affiliation(s)
- Cameron W Evans
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Diwei Ho
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Joshua B Marlow
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jessica J King
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Charmaine Hee
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Lucas N Wong
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Rob Atkin
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Nicole M Smith
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - Gregory G Warr
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Marck Norret
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| | - K Swaminathan Iyer
- School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia
| |
Collapse
|
28
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
Atz Dick T, Uludağ H. A Polyplex in a Shell: The Effect of Poly(aspartic acid)-Mediated Calcium Carbonate Mineralization on Polyplexes Properties and Transfection Efficiency. Mol Pharm 2022; 19:2077-2091. [PMID: 35649175 DOI: 10.1021/acs.molpharmaceut.1c00909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mineralization by exposure of organic templates to supersaturated solutions is used by many living organisms to generate specialized materials to perform structural or protective functions. Similarly, it was suggested that improved robustness acquired through mineralization under natural conditions could be an important factor for virus survival outside of a host for better transfection of cells. Here, inspired by this fact, we developed a nonviral tricomponent polyplex system for gene delivery capable of undergoing mineralization. First, we fabricated anionic polyplexes carrying pDNA by self-assembly with a lipid-modified cationic polymer and coating by poly(aspartic acid). Then, we submitted the polyplexes to a two-step mineralization reaction to precipitate CaCO3 under various supersaturations. We carried out detailed morphological studies of the mineralized polyplexes and identified which parameters of the fabrication process were influential on transfection efficiency. We found that mineralization with CaCO3 is efficient in promoting transfection efficiency as long as a certain Ca2+/CO32- lower limit ratio is respected. However, calcium incubation can also be used to achieve similar effects at higher concentrations depending on polyplex composition, probably due to the formation of physical cross-links by calcium binding to poly(aspartic acid). We proposed that the improved robustness and transfection efficiency provided by means of mineralization can be used to expand the possible applications of polyplexes in gene therapy.
Collapse
Affiliation(s)
- Teo Atz Dick
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T5K 2Y3 Canada
| | - Hasan Uludağ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T5K 2Y3 Canada.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3 Canada.,Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
30
|
Shupe J, Zhang A, Odenwelder DC, Dobrowsky T. Gene therapy: challenges in cell culture scale-up. Curr Opin Biotechnol 2022; 75:102721. [DOI: 10.1016/j.copbio.2022.102721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/04/2022] [Accepted: 03/02/2022] [Indexed: 11/03/2022]
|
31
|
Dick TA, Sone ED, Uludağ H. Mineralized vectors for gene therapy. Acta Biomater 2022; 147:1-33. [PMID: 35643193 DOI: 10.1016/j.actbio.2022.05.036] [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: 12/02/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/01/2022]
Abstract
There is an intense interest in developing materials for safe and effective delivery of polynucleotides using non-viral vectors. Mineralization of organic templates has long been used to produce complex materials with outstanding biocompatibility. However, a lack of control over mineral growth has limited the applicability of mineralized materials to a few in vitro applications. With better control over mineral growth and surface functionalization, mineralized vectors have advanced significantly in recent years. Here, we review the recent progress in chemical synthesis, physicochemical properties, and applications of mineralized materials in gene therapy, focusing on structure-function relationships. We contrast the classical understanding of the mineralization mechanism with recent ideas of mineralization. A brief introduction to gene delivery is summarized, followed by a detailed survey of current mineralized vectors. The vectors derived from calcium phosphate are articulated and compared to other minerals with unique features. Advanced mineral vectors derived from templated mineralization and specialty coatings are critically analyzed. Mineral systems beyond the co-precipitation are explored as more complex multicomponent systems. Finally, we conclude with a perspective on the future of mineralized vectors by carefully demarcating the boundaries of our knowledge and highlighting ambiguous areas in mineralized vectors. STATEMENT OF SIGNIFICANCE: Therapy by gene-based medicines is increasingly utilized to cure diseases that are not alleviated by conventional drug therapy. Gene medicines, however, rely on macromolecular nucleic acids that are too large and too hydrophilic for cellular uptake. Without tailored materials, they are not functional for therapy. One emerging class of nucleic acid delivery system is mineral-based materials. The fact that they can undergo controlled dissolution with minimal footprint in biological systems are making them attractive for clinical use, where safety is utmost importance. In this submission, we will review the emerging synthesis technology and the range of new generation minerals for use in gene medicines.
Collapse
|
32
|
Random Copolymers of Lysine and Isoleucine for Efficient mRNA Delivery. Int J Mol Sci 2022; 23:ijms23105363. [PMID: 35628177 PMCID: PMC9140483 DOI: 10.3390/ijms23105363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
Messenger RNA (mRNA) is currently of great interest as a new category of therapeutic agent, which could be used for prevention or treatment of various diseases. For this mRNA requires effective delivery systems that will protect it from degradation, as well as allow cellular uptake and mRNA release. Random poly(lysine-co-isoleucine) polypeptides were synthesized and investigated as possible carriers for mRNA delivery. The polypeptides obtained under lysine:isoleucine monomer ratio equal to 80/20 were shown to give polyplexes with smaller size, positive ζ-potential and more than 90% encapsulation efficacy. The phase inversion method was proposed as best way for encapsulation of mRNA into polyplexes, which are based on obtained amphiphilic copolymers. These copolymers showed efficacy in protection of bound mRNA towards ribonuclease and lower toxicity as compared to lysine homopolymer. The poly(lysine-co-isoleucine) polypeptides showed greater than poly(ethyleneimine) efficacy as vectors for transfection of cells with green fluorescent protein and firefly luciferase encoding mRNAs. This allows us to consider obtained copolymers as promising candidates for mRNA delivery applications.
Collapse
|
33
|
Elzes MR, Mertens I, Sedlacek O, Verbraeken B, Doensen ACA, Mees MA, Glassner M, Jana S, Paulusse JMJ, Hoogenboom R. Linear Poly(ethylenimine-propylenimine) Random Copolymers for Gene Delivery: From Polymer Synthesis to Efficient Transfection with High Serum Tolerance. Biomacromolecules 2022; 23:2459-2470. [PMID: 35499242 DOI: 10.1021/acs.biomac.2c00210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Naturally occurring oligoamines, such as spermine, spermidine, and putrescine, are well-known regulators of gene expression. These oligoamines frequently have short alkyl spacers with varying lengths between the amines. Linear polyethylenimine (PEI) is a polyamine that has been widely applied as a gene vector, with various formulations currently in clinical trials. In order to emulate natural oligoamine gene regulators, linear random copolymers containing both PEI and polypropylenimine (PPI) repeat units were designed as novel gene delivery agents. In general, statistical copolymerization of 2-oxazolines and 2-oxazines leads to the formation of gradient copolymers. In this study, however, we describe for the first time the synthesis of near-ideal random 2-oxazoline/2-oxazine copolymers through careful tuning of the monomer structures and reactivity as well as polymerization conditions. These copolymers were then transformed into near-random PEI-PPI copolymers by controlled side-chain hydrolysis. The prepared PEI-PPI copolymers formed stable polyplexes with GFP-encoding plasmid DNA, as validated by dynamic light scattering. Furthermore, the cytotoxicity and transfection efficiency of polyplexes were evaluated in C2C12 mouse myoblasts. While the polymer chain length did not significantly increase the toxicity, a higher PPI content was associated with increased toxicity and also lowered the amount of polymers needed to achieve efficient transfection. The transfection efficiency was significantly influenced by the degree of polymerization of PEI-PPI, whereby longer polymers resulted in more transfected cells. Copolymers with 60% or lower PPI content exhibited a good balance between high plasmid-DNA transfection efficiency and low toxicity. Interestingly, these novel PEI-PPI copolymers revealed exceptional serum tolerance, whereby transfection efficiencies of up to 53% of transfected cells were achieved even under 50% serum conditions. These copolymers, especially PEI-PPI with DP500 and a 1:1 PEI/PPI ratio, were identified as promising transfection agents for plasmid DNA.
Collapse
Affiliation(s)
- M Rachèl Elzes
- Department of Biomolecular Nanotechnology, MESA + Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, 7500 AE Enschede, The Netherlands
| | - Ine Mertens
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4, 9000 Ghent, Belgium
| | - Ondrej Sedlacek
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4, 9000 Ghent, Belgium.,Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, 12800 Prague, Czech Republic
| | - Bart Verbraeken
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4, 9000 Ghent, Belgium
| | - Aniek C A Doensen
- Department of Biomolecular Nanotechnology, MESA + Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, 7500 AE Enschede, The Netherlands.,Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4, 9000 Ghent, Belgium
| | - Maarten A Mees
- Department of Biomolecular Nanotechnology, MESA + Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, 7500 AE Enschede, The Netherlands
| | - Mathias Glassner
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4, 9000 Ghent, Belgium
| | - Somdeb Jana
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4, 9000 Ghent, Belgium
| | - Jos M J Paulusse
- Department of Biomolecular Nanotechnology, MESA + Institute for Nanotechnology and TechMed Institute for Health and Biomedical Technologies, Faculty of Science and Technology, University of Twente, 7500 AE Enschede, The Netherlands
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4, 9000 Ghent, Belgium
| |
Collapse
|
34
|
Kurt E, Segura T. Nucleic Acid Delivery from Granular Hydrogels. Adv Healthc Mater 2022; 11:e2101867. [PMID: 34742164 PMCID: PMC8810690 DOI: 10.1002/adhm.202101867] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/29/2021] [Indexed: 02/03/2023]
Abstract
Nucleic acid delivery has applications ranging from tissue engineering to vaccine development to infectious disease. Cationic polymer condensed nucleic acids are used with surface-coated porous scaffolds and are able to promote long-term gene expression. However, due to surface loading of the scaffold, there is a limit to the amount of nucleic acid that can be loaded, resulting in decreasing expression rate over time. In addition, surface-coated scaffolds are generally non-injectable. Here, it is demonstrated that cationic polymer condensed nucleic acids can be effectively loaded into injectable granular hydrogel scaffolds by stabilizing the condensed nucleic acid into a lyophilized powder, loading the powder into a bulk hydrogel, and then fragmenting the loaded hydrogel. The resulting hydrogel microparticles contain non-aggregated nucleic acid particles, can be annealed post-injection to result in an injectable microporous hydrogel, and can effectively deliver nucleic acids to embedded cells with a constant expression rate. Due to the nature of granular hydrogels, it is demonstrated that mixtures of loaded and unloaded particles and spatially resolved gene expression can be easily achieved. The ability to express genes long term from an injectable porous hydrogel will further open the applications of nucleic acid delivery.
Collapse
Affiliation(s)
- Evan Kurt
- Department of Biomedical Engineering, Duke University, Durham, NC
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC
- Departments Neurology and Dermatology, Duke University, Durham, NC
| |
Collapse
|
35
|
González-Domínguez I, Puente-Massaguer E, Lavado-García J, Cervera L, Gòdia F. Micrometric DNA/PEI polyplexes correlate with higher transient gene expression yields in HEK 293 cells. N Biotechnol 2022; 68:87-96. [DOI: 10.1016/j.nbt.2022.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/25/2022]
|
36
|
Coelho F, Salonen LM, Silva BFB. Hemiacetal-linked pH-sensitive PEG-lipids for non-viral gene delivery. NEW J CHEM 2022. [DOI: 10.1039/d2nj02217f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cationic lipid–DNA complexes containing a novel hemiacetal PEG-lipid for endosomal escape were characterized in terms of pH-response, stability, and biological activity.
Collapse
Affiliation(s)
- Filipe Coelho
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Laura M. Salonen
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Bruno F. B. Silva
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| |
Collapse
|
37
|
Tortajada L, Felip C, Vicent MJ. Polymer-based Non-viral Vectors for Gene Therapy in the Skin. Polym Chem 2022. [DOI: 10.1039/d1py01485d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gene therapy has emerged as a versatile technique with the potential to treat a range of human diseases; however, examples of the topical application of gene therapy as a treatment...
Collapse
|
38
|
García-Garrido E, Cordani M, Somoza Á. Modified Gold Nanoparticles to Overcome the Chemoresistance to Gemcitabine in Mutant p53 Cancer Cells. Pharmaceutics 2021; 13:2067. [PMID: 34959348 PMCID: PMC8703659 DOI: 10.3390/pharmaceutics13122067] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 12/29/2022] Open
Abstract
Mutant p53 proteins result from missense mutations in the TP53 gene, the most mutated in human cancer, and have been described to contribute to cancer initiation and progression. Therapeutic strategies for targeting mutant p53 proteins in cancer cells are limited and have proved unsuitable for clinical application due to problems related to drug delivery and toxicity to healthy tissues. Therefore, the discovery of efficient and safe therapeutic strategies that specifically target mutant p53 remains challenging. In this study, we generated gold nanoparticles (AuNPs) chemically modified with low molecular branched polyethylenimine (bPEI) for the efficient delivery of gapmers targeting p53 mutant protein. The AuNPs formulation consists of a combination of polymeric mixed layer of polyethylene glycol (PEG) and PEI, and layer-by-layer assembly of bPEI through a sensitive linker. These nanoparticles can bind oligonucleotides through electrostatic interactions and release them in the presence of a reducing agent as glutathione. The nanostructures generated here provide a non-toxic and powerful system for the delivery of gapmers in cancer cells, which significantly downregulated mutant p53 proteins and altered molecular markers related to cell growth and apoptosis, thus overcoming chemoresistance to gemcitabine.
Collapse
Affiliation(s)
- Eduardo García-Garrido
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
| | - Marco Cordani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Faraday 9, 28049 Madrid, Spain
- Unidad Asociada al Centro Nacional de Biotecnología (CSIC), Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
39
|
Alazzo A, Gumus N, Gurnani P, Stolnik S, Rahman R, Spriggs K, Alexander C. Investigating histidinylated highly branched poly(lysine) for siRNA delivery. J Mater Chem B 2021; 10:236-246. [PMID: 34852030 DOI: 10.1039/d1tb01793d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The temporary silencing of disease-associated genes utilising short interfering RNA (siRNA) is a potent and selective route for addressing a wide range of life limiting disorders. However, the few clinically approved siRNA therapies rely on lipid based formulations, which although potent, provide limited chemical space to tune the stability, efficacy and tissue selectivity. In this study, we investigated the role of molar mass and histidinylation for poly(lysine) based non-viral vectors, synthesised through a fully aqueous thermal condensation polymerisation. Formulation and in vitro studies revealed that higher molar mass derivatives yielded smaller polyplexes attributed to a greater affinity for siRNA at lower N/P ratios yielding greater transfection efficiency, albeit with some cytotoxicity. Histidinylation had a negligible effect on formulation size, yet imparted a moderate improvement in biocompatibility, but did not provide any meaningful improvement over silencing efficiency compared to non-histidinylated derivatives. This was attributed to a greater degree of cellular internalisation for non-histidinylated analogues, which was enhanced with the higher molar mass material.
Collapse
Affiliation(s)
- Ali Alazzo
- Department of Pharmaceutics, College of Pharmacy, University of Mosul, Mosul, Iraq.,Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Nurcan Gumus
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Pratik Gurnani
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Snjezana Stolnik
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Ruman Rahman
- BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Keith Spriggs
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
40
|
Coelho F, Botelho C, Paris JL, Marques EF, Silva BF. Influence of the media ionic strength on the formation and in vitro biological performance of polycation-DNA complexes. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.117930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
41
|
Garland KM, Rosch JC, Carson CS, Wang-Bishop L, Hanna A, Sevimli S, Van Kaer C, Balko JM, Ascano M, Wilson JT. Pharmacological Activation of cGAS for Cancer Immunotherapy. Front Immunol 2021; 12:753472. [PMID: 34899704 PMCID: PMC8662543 DOI: 10.3389/fimmu.2021.753472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/29/2021] [Indexed: 01/23/2023] Open
Abstract
When compartmentally mislocalized within cells, nucleic acids can be exceptionally immunostimulatory and can even trigger the immune-mediated elimination of cancer. Specifically, the accumulation of double-stranded DNA in the cytosol can efficiently promote antitumor immunity by activating the cGAMP synthase (cGAS) / stimulator of interferon genes (STING) cellular signaling pathway. Targeting this cytosolic DNA sensing pathway with interferon stimulatory DNA (ISD) is therefore an attractive immunotherapeutic strategy for the treatment of cancer. However, the therapeutic activity of ISD is limited by several drug delivery barriers, including susceptibility to deoxyribonuclease degradation, poor cellular uptake, and inefficient cytosolic delivery. Here, we describe the development of a nucleic acid immunotherapeutic, NanoISD, which overcomes critical delivery barriers that limit the activity of ISD and thereby promotes antitumor immunity through the pharmacological activation of cGAS at the forefront of the STING pathway. NanoISD is a nanoparticle formulation that has been engineered to confer deoxyribonuclease resistance, enhance cellular uptake, and promote endosomal escape of ISD into the cytosol, resulting in potent activation of the STING pathway via cGAS. NanoISD mediates the local production of proinflammatory cytokines via STING signaling. Accordingly, the intratumoral administration of NanoISD induces the infiltration of natural killer cells and T lymphocytes into murine tumors. The therapeutic efficacy of NanoISD is demonstrated in preclinical tumor models by attenuated tumor growth, prolonged survival, and an improved response to immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Kyle M. Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Jonah C. Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Carcia S. Carson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Lihong Wang-Bishop
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Ann Hanna
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sema Sevimli
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Casey Van Kaer
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Justin M. Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Manuel Ascano
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, United States
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
42
|
Diaz IL, Jérôme V, Freitag R, Perez LD. Development of poly(ethyleneimine) grafted amphiphilic copolymers: Evaluation of their cytotoxicity and ability to complex DNA. J BIOACT COMPAT POL 2021. [DOI: 10.1177/08839115211053925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Poly(ethyleneimine) (PEI) is one of the most widely used cationic polymers for gene delivery. The high molecular weight polymer, which is commercially available, is highly efficient but also very cytotoxic. The reduction in charge density by using nonlinear architectures based on low molecular weight (LMW) PEI is a promising approach to produce safer DNA-vectors. Herein, a group of cationic graft copolymers with different composition containing a hydrophobic biocompatible backbone and LMW linear PEI (lPEI) grafts obtained by ring opening polymerization and click chemistry was studied. The self-assembly and DNA complexation behavior of these materials was analyzed by the gel retardation assay, zeta potential measurements, and dynamic light scattering. The copolymers formed positively charged particles in water with average sizes between 270 and 377 nm. After they were added to DNA in serum-free medium, these particles acquired negative/near-neutral charges and increased in size depending on the N/P ratio. All copolymers showed reduced cytotoxicity compared to the 25 kDa lPEI used as reference, but the transfection efficiency was reduced. This result suggested that the cationic segments were too small to fully condense the DNA and promote cellular uptake, even with the use of several grafts and the introduction of hydrophobic domains. The trends found in this research showed that a higher degree of hydrophobicity and a higher grafting density can enhance the interaction between the copolymers and DNA. These trends could direct further structural modifications in the search for effective and safe vectors based on this polycation.
Collapse
Affiliation(s)
- Ivonne L Diaz
- Departamento de Química, Universidad Nacional de Colombia, Bogotá DC, Colombia
| | - Valérie Jérôme
- Process Biotechnology, University of Bayreuth, Bayreuth, Germany
| | - Ruth Freitag
- Process Biotechnology, University of Bayreuth, Bayreuth, Germany
| | - León D Perez
- Departamento de Química, Universidad Nacional de Colombia, Bogotá DC, Colombia
| |
Collapse
|
43
|
A Dick T, Uludağ H. Mineralized polyplexes for gene delivery: Improvement of transfection efficiency as a consequence of calcium incubation and not mineralization. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 129:112419. [PMID: 34579928 DOI: 10.1016/j.msec.2021.112419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 12/18/2022]
Abstract
Gene therapy is an emerging field in which nucleic acids are used to control protein expression. The necessity of delivering nucleic acids to specific cell types and intracellular sites demands the use of highly specialized gene carriers. As a carrier modification technique, mineralization has been successfully used to modify viral and non-viral carriers, providing new properties that ultimately aim to increase the transfection efficiency. However, for the specific case of polyplexes used in gene therapy, recent literature shows that interaction with calcium, a fundamental step of mineralization, might be effective to increase transfection efficiency, leaving an ambiguity about of the role of mineralization for this type of gene carriers. To answer this question and to reveal the properties responsible for increasing transfection efficiency, we mineralized poly(aspartic acid) coated polyplexes at various CaCl2 and Na3PO4 concentrations, and evaluated the resultant carriers for physicochemical and morphological characteristics, as well as transfection and delivery efficiency with MC3T3-E1 mouse osteoblastic cells. We found that both mineralization and calcium incubation positively affected the transfection efficiency and uptake of polyplexes in MC3T3-E1 cells. However, this effect originated from the properties achieved by polyplexes after the calcium incubation step that are maintained after mineralization, including particle size increase, improved pDNA binding, and adjustment of zeta potential. Considering that mineralization can be a longer process than calcium incubation, we find that calcium incubation might be sufficient and preferred if improved transfection efficiency in vitro is the only effect desired.
Collapse
Affiliation(s)
- Teo A Dick
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada.
| | - Hasan Uludağ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada; Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
44
|
Lin MHC, Lai PS, Chang LC, Huang WC, Lee MH, Chen KT, Chung CY, Yang JT. Characterization and Optimization of Chitosan-Coated Polybutylcyanoacrylate Nanoparticles for the Transfection-Guided Neural Differentiation of Mouse Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:8741. [PMID: 34445447 PMCID: PMC8395893 DOI: 10.3390/ijms22168741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/07/2021] [Accepted: 08/11/2021] [Indexed: 01/04/2023] Open
Abstract
Gene transfection is a valuable tool for analyzing gene regulation and function, and providing an avenue for the genetic engineering of cells for therapeutic purposes. Though efficient, the potential concerns over viral vectors for gene transfection has led to research in non-viral alternatives. Cationic polyplexes such as those synthesized from chitosan offer distinct advantages such as enhanced polyplex stability, cellular uptake, endo-lysosomal escape, and release, but are limited by the poor solubility and viscosity of chitosan. In this study, the easily synthesized biocompatible and biodegradable polymeric polysorbate 80 polybutylcyanoacrylate nanoparticles (PS80 PBCA NP) are utilized as the backbone for surface modification with chitosan, in order to address the synthetic issues faced when using chitosan alone as a carrier. Plasmid DNA (pDNA) containing the brain-derived neurotrophic factor (BDNF) gene coupled to a hypoxia-responsive element and the cytomegalovirus promotor gene was selected as the genetic cargo for the in vitro transfection-guided neural-lineage specification of mouse induced pluripotent stem cells (iPSCs), which were assessed by immunofluorescence staining. The chitosan-coated PS80 PBCA NP/BDNF pDNA polyplex measured 163.8 ± 1.8 nm and zeta potential measured -34.8 ± 1.8 mV with 0.01% (w/v) high molecular weight chitosan (HMWC); the pDNA loading efficiency reached 90% at a nanoparticle to pDNA weight ratio of 15, which also corresponded to enhanced polyplex stability on the DNA stability assay. The HMWC-PS80 PBCA NP/BDNF pDNA polyplex was non-toxic to mouse iPSCs for up to 80 μg/mL (weight ratio = 40) and enhanced the expression of BDNF when compared with PS80 PBCA NP/BDNF pDNA polyplex. Evidence for neural-lineage specification of mouse iPSCs was observed by an increased expression of nestin, neurofilament heavy polypeptide, and beta III tubulin, and the effects appeared superior when transfection was performed with the chitosan-coated formulation. This study illustrates the versatility of the PS80 PBCA NP and that surface decoration with chitosan enabled this delivery platform to be used for the transfection-guided differentiation of mouse iPSCs.
Collapse
Affiliation(s)
- Martin Hsiu-Chu Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan;
- PhD Programme of Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan;
| | - Li-Ching Chang
- Department of Dentistry, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan;
- Department of Nursing, Chang Gung University of Science and Technology, Chia-Yi 61363, Taiwan
| | - Wei-Chao Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Ming-Hsueh Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Kuo-Tai Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Chiu-Yen Chung
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
| | - Jen-Tsung Yang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Chia-Yi Branch, Chia-Yi 61363, Taiwan; (M.H.-C.L.); (W.-C.H.); (M.-H.L.); (K.-T.C.)
- College of Medicine, School of Traditional Chinese Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan
| |
Collapse
|
45
|
O'Keeffe Ahern J, Lara-Sáez I, Zhou D, Murillas R, Bonafont J, Mencía Á, García M, Manzanares D, Lynch J, Foley R, Xu Q, Sigen A, Larcher F, Wang W. Non-viral delivery of CRISPR-Cas9 complexes for targeted gene editing via a polymer delivery system. Gene Ther 2021; 29:157-170. [PMID: 34363036 PMCID: PMC9013665 DOI: 10.1038/s41434-021-00282-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022]
Abstract
Recent advances in molecular biology have led to the CRISPR revolution, but the lack of an efficient and safe delivery system into cells and tissues continues to hinder clinical translation of CRISPR approaches. Polymeric vectors offer an attractive alternative to viruses as delivery vectors due to their large packaging capacity and safety profile. In this paper, we have demonstrated the potential use of a highly branched poly(β-amino ester) polymer, HPAE-EB, to enable genomic editing via CRISPRCas9-targeted genomic excision of exon 80 in the COL7A1 gene, through a dual-guide RNA sequence system. The biophysical properties of HPAE-EB were screened in a human embryonic 293 cell line (HEK293), to elucidate optimal conditions for efficient and cytocompatible delivery of a DNA construct encoding Cas9 along with two RNA guides, obtaining 15–20% target genomic excision. When translated to human recessive dystrophic epidermolysis bullosa (RDEB) keratinocytes, transfection efficiency and targeted genomic excision dropped. However, upon delivery of CRISPR–Cas9 as a ribonucleoprotein complex, targeted genomic deletion of exon 80 was increased to over 40%. Our study provides renewed perspective for the further development of polymer delivery systems for application in the gene editing field in general, and specifically for the treatment of RDEB.
Collapse
Affiliation(s)
| | - Irene Lara-Sáez
- Charles Institute of Dermatology, University College Dublin, Dublin, Republic of Ireland.
| | - Dezhong Zhou
- Charles Institute of Dermatology, University College Dublin, Dublin, Republic of Ireland
| | - Rodolfo Murillas
- Epithelial Biomedicine Division, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain.,Fundación Instituto de Investigaciones Sanitarias de la Fundación Jimenez Díaz, Madrid, Spain
| | - Jose Bonafont
- Epithelial Biomedicine Division, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain.,Fundación Instituto de Investigaciones Sanitarias de la Fundación Jimenez Díaz, Madrid, Spain
| | - Ángeles Mencía
- Epithelial Biomedicine Division, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Marta García
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain.,Fundación Instituto de Investigaciones Sanitarias de la Fundación Jimenez Díaz, Madrid, Spain.,Department of Bioengineering Universidad Carlos III de Madrid, Madrid, Spain
| | - Darío Manzanares
- Charles Institute of Dermatology, University College Dublin, Dublin, Republic of Ireland
| | - Jennifer Lynch
- Charles Institute of Dermatology, University College Dublin, Dublin, Republic of Ireland
| | - Ruth Foley
- Charles Institute of Dermatology, University College Dublin, Dublin, Republic of Ireland
| | - Qian Xu
- Charles Institute of Dermatology, University College Dublin, Dublin, Republic of Ireland
| | - A Sigen
- Charles Institute of Dermatology, University College Dublin, Dublin, Republic of Ireland
| | - Fernando Larcher
- Epithelial Biomedicine Division, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain.,Fundación Instituto de Investigaciones Sanitarias de la Fundación Jimenez Díaz, Madrid, Spain.,Department of Bioengineering Universidad Carlos III de Madrid, Madrid, Spain
| | - Wenxin Wang
- Charles Institute of Dermatology, University College Dublin, Dublin, Republic of Ireland.
| |
Collapse
|
46
|
Cortez‐Jugo C, Czuba‐Wojnilowicz E, Tan A, Caruso F. A Focus on "Bio" in Bio-Nanoscience: The Impact of Biological Factors on Nanomaterial Interactions. Adv Healthc Mater 2021; 10:e2100574. [PMID: 34170631 DOI: 10.1002/adhm.202100574] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/18/2021] [Indexed: 12/17/2022]
Abstract
Bio-nanoscience research encompasses studies on the interactions of nanomaterials with biological structures or what is commonly referred to as the biointerface. Fundamental studies on the influence of nanomaterial properties, including size, shape, composition, and charge, on the interaction with the biointerface have been central in bio-nanoscience to assess nanomaterial efficacy and safety for a range of biomedical applications. However, the state of the cells, tissues, or biological models can also influence the behavior of nanomaterials at the biointerface and their intracellular processing. Focusing on the "bio" in bio-nano, this review discusses the impact of biological properties at the cellular, tissue, and whole organism level that influences nanomaterial behavior, including cell type, cell cycle, tumor physiology, and disease states. Understanding how the biological factors can be addressed or exploited to enhance nanomaterial accumulation and uptake can guide the design of better and suitable models to improve the outcomes of materials in nanomedicine.
Collapse
Affiliation(s)
- Christina Cortez‐Jugo
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Ewa Czuba‐Wojnilowicz
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Abigail Tan
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering The University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
47
|
Liu HW, Hu Y, Ren Y, Nam H, Santos JL, Ng S, Gong L, Brummet M, Carrington CA, Ullman CG, Pomper MG, Minn I, Mao HQ. Scalable Purification of Plasmid DNA Nanoparticles by Tangential Flow Filtration for Systemic Delivery. ACS APPLIED MATERIALS & INTERFACES 2021; 13:30326-30336. [PMID: 34162211 PMCID: PMC9701136 DOI: 10.1021/acsami.1c05750] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Plasmid DNA (pDNA) nanoparticles synthesized by complexation with linear polyethylenimine (lPEI) are one of the most effective non-viral gene delivery vehicles. However, the lack of scalable and reproducible production methods and the high toxicity have hindered their clinical translation. Previously, we have developed a scalable flash nanocomplexation (FNC) technique to formulate pDNA/lPEI nanoparticles using a continuous flow process. Here, we report a tangential flow filtration (TFF)-based scalable purification method to reduce the uncomplexed lPEI concentration in the nanoparticle formulation and improve its biocompatibility. The optimized procedures achieved a 60% reduction of the uncomplexed lPEI with preservation of the nanoparticle size and morphology. Both in vitro and in vivo studies showed that the purified nanoparticles significantly reduced toxicity while maintaining transfection efficiency. TFF also allows for gradual exchange of solvents to isotonic solutions and further concentrating the nanoparticles for injection. Combining FNC production and TFF purification, we validated the purified pDNA/lPEI nanoparticles for future clinical translation of this gene nanomedicine.
Collapse
Affiliation(s)
- Heng-Wen Liu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yizong Hu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yong Ren
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hwanhee Nam
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jose Luis Santos
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Shirley Ng
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Like Gong
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Mary Brummet
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | - Martin G. Pomper
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Il Minn
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
48
|
Zu H, Gao D. Non-viral Vectors in Gene Therapy: Recent Development, Challenges, and Prospects. AAPS JOURNAL 2021; 23:78. [PMID: 34076797 PMCID: PMC8171234 DOI: 10.1208/s12248-021-00608-7] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/07/2021] [Indexed: 12/16/2022]
Abstract
Gene therapy has been experiencing a breakthrough in recent years, targeting various specific cell groups in numerous therapeutic areas. However, most recent clinical studies maintain the use of traditional viral vector systems, which are challenging to manufacture cost-effectively at a commercial scale. Non-viral vectors have been a fast-paced research topic in gene delivery, such as polymers, lipids, inorganic particles, and combinations of different types. Although non-viral vectors are low in their cytotoxicity, immunogenicity, and mutagenesis, attracting more and more researchers to explore the promising delivery system, they do not carry ideal characteristics and have faced critical challenges, including gene transfer efficiency, specificity, gene expression duration, and safety. This review covers the recent advancement in non-viral vectors research and formulation aspects, the challenges, and future perspectives.
Collapse
Affiliation(s)
- Hui Zu
- Abbvie Inc., 1 N. Waukegan Rd, North Chicago, Illinois, 60064, USA
| | - Danchen Gao
- Abbvie Inc., 1 N. Waukegan Rd, North Chicago, Illinois, 60064, USA.
| |
Collapse
|
49
|
Annenkov VV, Danilovtseva EN, Zelinskiy SN, Pal'shin VA. Submicro- and nanoplastics: How much can be expected in water bodies? ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 278:116910. [PMID: 33743272 DOI: 10.1016/j.envpol.2021.116910] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/17/2021] [Accepted: 03/08/2021] [Indexed: 06/12/2023]
Abstract
Plastic particles smaller than 1 μm are considered to be highly dangerous pollutants due to their ability to penetrate living cells. Model experiments on the toxicity of plastics should be correlated with actual concentrations of plastics in natural water. We simulated the natural destruction of polystyrene, polyvinyl chloride, and poly(methyl methacrylate) in experiments on the abrasion of plastics with small stones. The plastics were dyed in mass with a fluorescent dye, which made it possible to distinguish plastic particles from stone fragments. We found that less than 1% of polystyrene and polyvinyl chloride were converted to submicron size particles. In the case of more rigid poly(methyl methacrylate), the fraction of such particles reaches 11%. The concentration of particles with a diameter less than 1 μm in the model experiments was from 0.7 (polystyrene) to 13 mg/L (poly(methyl methacrylate)), and when transferring the obtained data to real reservoirs, these values should be reduced by several orders of magnitude. These data explain the difficulties associated with the search for nanoplastics in natural waters. The toxicity of such particles to hydrobionts in model experiments was detected for concentrations greater than 1 mg/L, which is unrealistic in nature. Detectable and toxic amounts of nano- and submicron plastic particles in living organisms can be expected only in the case of filter-feeding organisms, such as molluscs, krill, sponges, etc.
Collapse
Affiliation(s)
- Vadim V Annenkov
- Limnological Institute, Siberian Branch of the Russian Academy of Sciences, Ulan-Batorskaya Str., 3, Irkutsk, 664033, Russia.
| | - Elena N Danilovtseva
- Limnological Institute, Siberian Branch of the Russian Academy of Sciences, Ulan-Batorskaya Str., 3, Irkutsk, 664033, Russia
| | - Stanislav N Zelinskiy
- Limnological Institute, Siberian Branch of the Russian Academy of Sciences, Ulan-Batorskaya Str., 3, Irkutsk, 664033, Russia
| | - Viktor A Pal'shin
- Limnological Institute, Siberian Branch of the Russian Academy of Sciences, Ulan-Batorskaya Str., 3, Irkutsk, 664033, Russia
| |
Collapse
|
50
|
N-[4-( N,N,N-Trimethylammonium)Benzyl]Chitosan Chloride as a Gene Carrier: The Influence of Polyplex Composition and Cell Type. MATERIALS 2021; 14:ma14092467. [PMID: 34068680 PMCID: PMC8126137 DOI: 10.3390/ma14092467] [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: 04/03/2021] [Revised: 04/24/2021] [Accepted: 05/06/2021] [Indexed: 11/16/2022]
Abstract
Polyplex-based gene delivery systems are promising substitutes for viral vectors because of their high versatility and lack of disadvantages commonly encountered with viruses. In this work, we studied the DNA polyplexes with N-[4-(N,N,N-trimethylammonium)benzyl]chitosan chloride (TMAB-CS) of various compositions in different cell types. Investigations of the interaction of TMAB-CS with DNA by different physical methods revealed that the molecular weight and the degree of substitution do not dramatically influence the hydrodynamic properties of polyplexes. Highly substituted TMAB-CS samples had a high affinity for DNA. The transfection protocol was optimized in HEK293T cells and achieved the highest efficiency of 30-35%. TMAB-CS was dramatically less effective in nonadherent K562 cells (around 1% transfected cells), but it was more effective and less toxic than polyarginine.
Collapse
|