1
|
Mm Yahya S, Elsayed GH. The role of MiRNA-34 family in different signaling pathways and its therapeutic options. Gene 2024; 931:148829. [PMID: 39154971 DOI: 10.1016/j.gene.2024.148829] [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: 01/19/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024]
Abstract
MiRNAs are short non-coding RNA molecules that have been shown to affect a vast number of genes at the post-transcriptional level, hence regulating several signaling pathways. Because the miRNA-34 family regulates a number of different signaling pathways, including those linked to cancer, the immune system, metabolism, cellular structure, and neurological disorders, it has garnered a great deal of attention from researchers. Members of the miRNA-34 family have been shown to inhibit tumors in a variety of cancer types. This family is also important for obesity, the cardiovascular system, and glycolysis. It's interesting to note that the miRNA-34 family is known to play a role in major depressive disorder, schizophrenia, Parkinson's disease (PD), adverse childhood experiences or trauma, regulation of stress responses, Alzheimer's disease (AD), and stress-related psychatric conditions. In this review, the expected targets of the miRNA-34 family are presented alongside the well-established targets identified by pathway analysis. Furthermore, the therapeutic potential of this miRNA family will be discussed.
Collapse
Affiliation(s)
- Shaymaa Mm Yahya
- Hormones Department, Medical Research and Clinical Studies Institute, and Stem Cell Lab, Centre of Excellence for Advanced SciencesNational Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt.
| | - Ghada H Elsayed
- Hormones Department, Medical Research and Clinical Studies Institute, and Stem Cell Lab, Centre of Excellence for Advanced SciencesNational Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| |
Collapse
|
2
|
Henríquez R, Muñoz-Barroso I. Viral vector- and virus-like particle-based vaccines against infectious diseases: A minireview. Heliyon 2024; 10:e34927. [PMID: 39144987 PMCID: PMC11320483 DOI: 10.1016/j.heliyon.2024.e34927] [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: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
To overcome the limitations of conventional vaccines, new platforms for vaccine design have emerged such as those based on viral vectors and virus-like particles (VLPs). Viral vector vaccines are highly efficient and the onset of protection is quick. Many recombinant vaccine candidates for humans are based on viruses belonging to different families such as Adenoviridae, Retroviridae, Paramyxoviridae, Rhabdoviridae, and Parvoviridae. Also, the first viral vector vaccine licensed for human vaccination was the Japanese encephalitis virus vaccine. Since then, several viral vectors have been approved for vaccination against the viruses of Lassa fever, Ebola, hepatitis B, hepatitis E, SARS-CoV-2, and malaria. VLPs are nanoparticles that mimic viral particles formed from the self-assembly of structural proteins and VLP-based vaccines against hepatitis B and E viruses, human papillomavirus, and malaria have been commercialized. As evidenced by the accelerated production of vaccines against COVID-19, these new approaches are important tools for vaccinology and for generating rapid responses against pathogens and emerging pandemic threats.
Collapse
Affiliation(s)
- Ruth Henríquez
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| | - Isabel Muñoz-Barroso
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| |
Collapse
|
3
|
Fischer J, Fedotova A, Jaki L, Sallard E, Erhardt A, Fuchs J, Ruzsics Z. Combining CRISPR-Cas-mediated terminal resolution with a novel genetic workflow to achieve high-diversity adenoviral libraries. Mol Ther Methods Clin Dev 2024; 32:101241. [PMID: 38585687 PMCID: PMC10995876 DOI: 10.1016/j.omtm.2024.101241] [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: 11/28/2023] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
While recombinant adenoviruses (rAds) are widely used in both laboratory and medical gene transfer, library-based applications using this vector platform are not readily available. Recently, we developed a new method, the CRISPR-Cas9 mediated in vivo terminal resolution aiding high-efficiency rescue of rAds from recombinant DNA. Here we report on a genetic workflow that allows construction of bacterial artificial chromosome-based rAd libraries reconstituted using highly efficient terminal resolution. We utilized frequent, pre-existing genomic sequences to allow the insertion of a selection marker, complementing two selected target sites into novel endonuclease recognition sites. In the second step, this selection marker is replaced with a transgene or mutation of interest via Gibson assembly. Our approach does not cause unwanted genomic off-target mutations while providing substantial flexibility for the site and nature of the genetic modification. This new genetic workflow, which we termed half site-directed fragment replacement (HFR) allows the introduction of more than 106 unique modifications into rAd encoding BACs using laboratory scale methodology. To demonstrate the power of HFR, we rescued barcoded viral vector libraries yielding a diversity of approximately 2.5 × 104 unique rAds per cm2 of transfected cell culture.
Collapse
Affiliation(s)
- Julian Fischer
- Institute of Virology, University Medical Center Freiburg, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
| | - Ariana Fedotova
- Institute of Virology, University Medical Center Freiburg, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
| | - Lena Jaki
- Institute of Virology, University Medical Center Freiburg, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
| | - Erwan Sallard
- Virology and Microbiology, Centre for Biomedical Education & Research (ZBAF), Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Anja Erhardt
- Virology and Microbiology, Centre for Biomedical Education & Research (ZBAF), Faculty of Health, Witten/Herdecke University, 58453 Witten, Germany
| | - Jonas Fuchs
- Institute of Virology, University Medical Center Freiburg, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
| | - Zsolt Ruzsics
- Institute of Virology, University Medical Center Freiburg, Medical Faculty, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
4
|
Jamour P, Jamali A, Langeroudi AG, Sharafabad BE, Abdoli A. Comparing chemical transfection, electroporation, and lentiviral vector transduction to achieve optimal transfection conditions in the Vero cell line. BMC Mol Cell Biol 2024; 25:15. [PMID: 38741034 PMCID: PMC11089686 DOI: 10.1186/s12860-024-00511-x] [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: 01/24/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Transfection is an important analytical method for studying gene expression in the cellular environment. There are some barriers to efficient DNA transfection in host cells, including circumventing the plasma membrane, escaping endosomal compartmentalization, autophagy, immune sensing pathways, and translocating the nuclear envelope. Therefore, it would be very useful to introduce an optimum transfection approach to achieve a high transfection efficiency in the Vero cell line. The aim of this study was to compare various transfection techniques and introduce a highly efficient method for gene delivery in Vero cells. METHODS In the current study, three transfection methods were used, including chemical transfection, electroporation, and lentiviral vector transduction, to obtain the optimum transfection conditions in the Vero cell line. Vero cells were cultured and transfected with chemical transfection reagents, electroporation, or HIV-1-based lentivectors under different experimental conditions. Transfection efficiency was assessed using flow cytometry and fluorescence microscopy to detect GFP-positive cells. RESULTS Among the tested methods, TurboFect™ chemical transfection exhibited the highest efficiency. Optimal transfection conditions were achieved using 1 µg DNA and 4 µL TurboFect™ in 6 × 104 Vero cells. CONCLUSION TurboFect™, a cationic polymer transfection reagent, demonstrated superior transfection efficiency in Vero cells compared with electroporation and lentivirus particles, and is the optimal choice for chemical transfection in the Vero cell line.
Collapse
Affiliation(s)
- Parisa Jamour
- Department of Hepatitis and HIV, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Abbas Jamali
- Department of Influenza and Other Respiratory Viruses, Pasteur Institute of Iran, Tehran, Iran
| | - Arash Ghalyanchi Langeroudi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Behrouz Ebadi Sharafabad
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Asghar Abdoli
- Department of Hepatitis and HIV, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Ou J, Tang Y, Xu J, Tucci J, Borys MC, Khetan A. Recent advances in upstream process development for production of recombinant adeno-associated virus. Biotechnol Bioeng 2024; 121:53-70. [PMID: 37691172 DOI: 10.1002/bit.28545] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 05/17/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023]
Abstract
Recombinant adeno-associated virus (rAAV) is rapidly emerging as the preferred delivery vehicle for gene therapies, with promising advantages in safety and efficacy. Key challenges in systemic in-vivo rAAV gene therapy applications are the gap in production capabilities versus potential market demand and complex production process. This review summarizes current available information on rAAV upstream manufacturing processes and proposed optimizations for production. The advancements in rAAV production media were reviewed with proposals to speed up the cell culture process development. Furthermore, major methods for genetic element delivery to host cells were summarized with their advantages, limitations, and future directions for optimization. In addition, culture vessel selection criteria were listed based on production cell system, scale, and development stage. Process control at the production step was also outlined with an in-depth understanding of production kinetics and quality control.
Collapse
Affiliation(s)
- Jianfa Ou
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Yawen Tang
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Jianlin Xu
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Julian Tucci
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Michael C Borys
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| | - Anurag Khetan
- Biologics Development, Global Product Development and Supply, Bristol Myers Squibb, Devens, Massachusetts, USA
| |
Collapse
|
6
|
Perez-Sanchez J, Middleton SJ, Pattison LA, Hilton H, Awadelkareem MA, Zuberi SR, Renke MB, Hu H, Yang X, Clark AJ, Smith ESJ, Bennett DL. A humanized chemogenetic system inhibits murine pain-related behavior and hyperactivity in human sensory neurons. Sci Transl Med 2023; 15:eadh3839. [PMID: 37792955 PMCID: PMC7615191 DOI: 10.1126/scitranslmed.adh3839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Hyperexcitability in sensory neurons is known to underlie many of the maladaptive changes associated with persistent pain. Chemogenetics has shown promise as a means to suppress such excitability, yet chemogenetic approaches suitable for human applications are needed. PSAM4-GlyR is a modular system based on the human α7 nicotinic acetylcholine and glycine receptors, which responds to inert chemical ligands and the clinically approved drug varenicline. Here, we demonstrated the efficacy of this channel in silencing both mouse and human sensory neurons by the activation of large shunting conductances after agonist administration. Virally mediated expression of PSAM4-GlyR in mouse sensory neurons produced behavioral hyposensitivity upon agonist administration, which was recovered upon agonist washout. Stable expression of the channel led to similar reversible suppression of pain-related behavior even after 10 months of viral delivery. Mechanical and spontaneous pain readouts were also ameliorated by PSAM4-GlyR activation in acute and joint pain inflammation mouse models. Furthermore, suppression of mechanical hypersensitivity generated by a spared nerve injury model of neuropathic pain was also observed upon activation of the channel. Effective silencing of behavioral hypersensitivity was reproduced in a human model of hyperexcitability and clinical pain: PSAM4-GlyR activation decreased the excitability of human-induced pluripotent stem cell-derived sensory neurons and spontaneous activity due to a gain-of-function NaV1.7 mutation causing inherited erythromelalgia. Our results demonstrate the contribution of sensory neuron hyperexcitability to neuropathic pain and the translational potential of an effective, stable, and reversible humanized chemogenetic system for the treatment of pain.
Collapse
Affiliation(s)
- Jimena Perez-Sanchez
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Steven J. Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Luke A. Pattison
- Department of Pharmacology, University of Cambridge; Cambridge CB2 1PD, UK
| | - Helen Hilton
- Department of Pharmacology, University of Cambridge; Cambridge CB2 1PD, UK
| | | | - Sana R. Zuberi
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Maria B. Renke
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Huimin Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Xun Yang
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| | - Alex J. Clark
- Blizard Institute, Barts and the London School of Medicine and Dentistry; London E1 2AT, UK
| | | | - David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford OX3 9DU, UK
| |
Collapse
|
7
|
Reddy JV, Raudenbush K, Papoutsakis ET, Ierapetritou M. Cell-culture process optimization via model-based predictions of metabolism and protein glycosylation. Biotechnol Adv 2023; 67:108179. [PMID: 37257729 DOI: 10.1016/j.biotechadv.2023.108179] [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: 11/27/2022] [Revised: 05/18/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
In order to meet the rising demand for biologics and become competitive on the developing biosimilar market, there is a need for process intensification of biomanufacturing processes. Process development of biologics has historically relied on extensive experimentation to develop and optimize biopharmaceutical manufacturing. Experimentation to optimize media formulations, feeding schedules, bioreactor operations and bioreactor scale up is expensive, labor intensive and time consuming. Mathematical modeling frameworks have the potential to enable process intensification while reducing the experimental burden. This review focuses on mathematical modeling of cellular metabolism and N-linked glycosylation as applied to upstream manufacturing of biologics. We review developments in the field of modeling cellular metabolism of mammalian cells using kinetic and stoichiometric modeling frameworks along with their applications to simulate, optimize and improve mechanistic understanding of the process. Interest in modeling N-linked glycosylation has led to the creation of various types of parametric and non-parametric models. Most published studies on mammalian cell metabolism have performed experiments in shake flasks where the pH and dissolved oxygen cannot be controlled. Efforts to understand and model the effect of bioreactor-specific parameters such as pH, dissolved oxygen, temperature, and bioreactor heterogeneity are critically reviewed. Most modeling efforts have focused on the Chinese Hamster Ovary (CHO) cells, which are most commonly used to produce monoclonal antibodies (mAbs). However, these modeling approaches can be generalized and applied to any mammalian cell-based manufacturing platform. Current and potential future applications of these models for Vero cell-based vaccine manufacturing, CAR-T cell therapies, and viral vector manufacturing are also discussed. We offer specific recommendations for improving the applicability of these models to industrially relevant processes.
Collapse
Affiliation(s)
- Jayanth Venkatarama Reddy
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA
| | - Katherine Raudenbush
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA; Delaware Biotechnology Institute, Department of Biological Sciences, University of Delaware, USA.
| | - Marianthi Ierapetritou
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716-3196, USA.
| |
Collapse
|
8
|
Rocamora F, Peralta AG, Shin S, Sorrentino J, Wu MYM, Toth EA, Fuerst TR, Lewis NE. Glycosylation shapes the efficacy and safety of diverse protein, gene and cell therapies. Biotechnol Adv 2023; 67:108206. [PMID: 37354999 PMCID: PMC11168894 DOI: 10.1016/j.biotechadv.2023.108206] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Over recent decades, therapeutic proteins have had widespread success in treating a myriad of diseases. Glycosylation, a near universal feature of this class of drugs, is a critical quality attribute that significantly influences the physical properties, safety profile and biological activity of therapeutic proteins. Optimizing protein glycosylation, therefore, offers an important avenue to developing more efficacious therapies. In this review, we discuss specific examples of how variations in glycan structure and glycoengineering impacts the stability, safety, and clinical efficacy of protein-based drugs that are already in the market as well as those that are still in preclinical development. We also highlight the impact of glycosylation on next generation biologics such as T cell-based cancer therapy and gene therapy.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Angelo G Peralta
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seunghyeon Shin
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - James Sorrentino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mina Ying Min Wu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric A Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
9
|
Armas F, Chandra F, Lee WL, Gu X, Chen H, Xiao A, Leifels M, Wuertz S, Alm EJ, Thompson J. Contextualizing Wastewater-Based surveillance in the COVID-19 vaccination era. ENVIRONMENT INTERNATIONAL 2023; 171:107718. [PMID: 36584425 PMCID: PMC9783150 DOI: 10.1016/j.envint.2022.107718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 06/17/2023]
Abstract
SARS-CoV-2 wastewater-based surveillance (WBS) offers a tool for cost-effective oversight of a population's infections. In the past two years, WBS has proven to be crucial for managing the pandemic across different geographical regions. However, the changing context of the pandemic due to high levels of COVID-19 vaccination warrants a closer examination of its implication towards SARS-CoV-2 WBS. Two main questions were raised: 1) Does vaccination cause shedding of viral signatures without infection? 2) Does vaccination affect the relationship between wastewater and clinical data? To answer, we review historical reports of shedding from viral vaccines in use prior to the COVID-19 pandemic including for polio, rotavirus, influenza and measles infection and provide a perspective on the implications of different COVID-19 vaccination strategies with regard to the potential shedding of viral signatures into the sewershed. Additionally, we reviewed studies that looked into the relationship between wastewater and clinical data and how vaccination campaigns could have affected the relationship. Finally, analyzing wastewater and clinical data from the Netherlands, we observed changes in the relationship concomitant with increasing vaccination coverage and switches in dominant variants of concern. First, that no vaccine-derived shedding is expected from the current commercial pipeline of COVID-19 vaccines that may confound interpretation of WBS data. Secondly, that breakthrough infections from vaccinated individuals contribute significantly to wastewater signals and must be interpreted in light of the changing dynamics of shedding from new variants of concern.
Collapse
Affiliation(s)
- Federica Armas
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Franciscus Chandra
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Wei Lin Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Xiaoqiong Gu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Hongjie Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore
| | - Amy Xiao
- Department of Biological Engineering, Massachusetts Institute of Technology, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology
| | - Mats Leifels
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | - Stefan Wuertz
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; School of Civil and Environmental Engineering, Nanyang Technological University, Singapore
| | - Eric J Alm
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Department of Biological Engineering, Massachusetts Institute of Technology, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Janelle Thompson
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; Asian School of the Environment, Nanyang Technological University, Singapore.
| |
Collapse
|
10
|
Aptamers Enhance Oncolytic Viruses' Antitumor Efficacy. Pharmaceutics 2022; 15:pharmaceutics15010151. [PMID: 36678780 PMCID: PMC9864469 DOI: 10.3390/pharmaceutics15010151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
Oncolytic viruses are highly promising for cancer treatment because they target and lyse tumor cells. These genetically engineered vectors introduce therapeutic or immunostimulatory genes into the tumor. However, viral therapy is not always safe and effective. Several problems are related to oncolytic viruses' targeted delivery to the tumor and immune system neutralization in the bloodstream. Cryoprotection and preventing viral particles from aggregating during storage are other critical issues. Aptamers, short RNA, or DNA oligonucleotides may help to crawl through this bottleneck. They are not immunogenic, are easily synthesized, can be chemically modified, and are not very demanding in storage conditions. It is possible to select an aptamer that specifically binds to any target cell, oncolytic virus, or molecule using the SELEX technology. This review comprehensively highlights the most important research and methodological approaches related to oncolytic viruses and nucleic acid aptamers. Here, we also analyze possible future research directions for combining these two methodologies to improve the effectiveness of cancer virotherapy.
Collapse
|
11
|
Tran MY, Kamen AA. Production of Lentiviral Vectors Using a HEK-293 Producer Cell Line and Advanced Perfusion Processing. Front Bioeng Biotechnol 2022; 10:887716. [PMID: 35774066 PMCID: PMC9237754 DOI: 10.3389/fbioe.2022.887716] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/24/2022] [Indexed: 12/03/2022] Open
Abstract
The field of lentiviral vector (LV) production continues to face challenges in large-scale manufacturing, specifically regarding producing enough vectors to meet the demand for treating patients as well as producing high and consistent quality of vectors for efficient dosing. Two areas of interest are the use of stable producer cell lines, which facilitates the scalability of LV production processes as well as making the process more reproducible and robust for clinical applications, and the search of a cell retention device scalable to industrial-size bioreactors. This manuscript investigates a stable producer cell line for producing LVs with GFP as the transgene at shake flask scale and demonstrates LV production at 3L bioreactor scale using the Tangential Flow Depth Filtration (TFDF) as a cell retention device in perfusion mode. Cumulative functional yields of 3.3 x 1011 and 3.9 x 1011 transducing units were achieved; the former over 6 days of LV production with 16.3 L of perfused media and the latter over 4 days with 16 L. In comparing to a previously published value that was achieved using the same stable producer cell line and the acoustic filter as the perfusion device at the same bioreactor scale, the TFDF perfusion run produced 1.5-fold higher cumulative functional yield. Given its scale-up potential, the TFDF is an excellent candidate to be further evaluated to determine optimized conditions that can ultimately support continuous manufacturing of LVs at large scale.
Collapse
|
12
|
Jt S, M H, Wam B, Ac B, Sa N. Adenoviral vectors for cardiovascular gene therapy applications: a clinical and industry perspective. J Mol Med (Berl) 2022; 100:875-901. [PMID: 35606652 PMCID: PMC9126699 DOI: 10.1007/s00109-022-02208-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 11/29/2022]
Abstract
Abstract Despite the development of novel pharmacological treatments, cardiovascular disease morbidity and mortality remain high indicating an unmet clinical need. Viral gene therapy enables targeted delivery of therapeutic transgenes and represents an attractive platform for tackling acquired and inherited cardiovascular diseases in the future. Current cardiovascular gene therapy trials in humans mainly focus on improving cardiac angiogenesis and function. Encouragingly, local delivery of therapeutic transgenes utilising first-generation human adenovirus serotype (HAd)-5 is safe in the short term and has shown some efficacy in drug refractory angina pectoris and heart failure with reduced ejection fraction. Despite this success, systemic delivery of therapeutic HAd-5 vectors targeting cardiovascular tissues and internal organs is limited by negligible gene transfer to target cells, elimination by the immune system, liver sequestration, off-target effects, and episomal degradation. To circumvent these barriers, cardiovascular gene therapy research has focused on determining the safety and efficacy of rare alternative serotypes and/or genetically engineered adenoviral capsid protein-modified vectors following local or systemic delivery. Pre-clinical studies have identified several vectors including HAd-11, HAd-35, and HAd-20–42-42 as promising platforms for local and systemic targeting of vascular endothelial and smooth muscle cells. In the past, clinical gene therapy trials were often restricted by limited scale-up capabilities of gene therapy medicinal products (GTMPs) and lack of regulatory guidance. However, significant improvement of industrial GTMP scale-up and purification, development of novel producer cell lines, and issuing of GTMP regulatory guidance by national regulatory health agencies have addressed many of these challenges, creating a more robust framework for future adenoviral-based cardiovascular gene therapy. In addition, this has enabled the mass roll out of adenovirus vector-based COVID-19 vaccines. Key messages First-generation HAd-5 vectors are widely used in cardiovascular gene therapy. HAd-5-based gene therapy was shown to lead to cardiac angiogenesis and improved function. Novel HAd vectors may represent promising transgene carriers for systemic delivery. Novel methods allow industrial scale-up of rare/genetically altered Ad serotypes. National regulatory health agencies have issued guidance on GMP for GTMPs.
Collapse
Affiliation(s)
- Schwartze Jt
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| | - Havenga M
- Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16, 2333, CL, Leiden, The Netherlands
| | - Bakker Wam
- Batavia Biosciences B.V., Bioscience Park Leiden, Zernikedreef 16, 2333, CL, Leiden, The Netherlands
| | - Bradshaw Ac
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Nicklin Sa
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
13
|
Casazza JP, Cale EM, Narpala S, Yamshchikov GV, Coates EE, Hendel CS, Novik L, Holman LA, Widge AT, Apte P, Gordon I, Gaudinski MR, Conan-Cibotti M, Lin BC, Nason MC, Trofymenko O, Telscher S, Plummer SH, Wycuff D, Adams WC, Pandey JP, McDermott A, Roederer M, Sukienik AN, O'Dell S, Gall JG, Flach B, Terry TL, Choe M, Shi W, Chen X, Kaltovich F, Saunders KO, Stein JA, Doria-Rose NA, Schwartz RM, Balazs AB, Baltimore D, Nabel GJ, Koup RA, Graham BS, Ledgerwood JE, Mascola JR. Safety and tolerability of AAV8 delivery of a broadly neutralizing antibody in adults living with HIV: a phase 1, dose-escalation trial. Nat Med 2022; 28:1022-1030. [PMID: 35411076 PMCID: PMC9876739 DOI: 10.1038/s41591-022-01762-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/28/2022] [Indexed: 01/27/2023]
Abstract
Adeno-associated viral vector-mediated transfer of DNA coding for broadly neutralizing anti-HIV antibodies (bnAbs) offers an alternative to attempting to induce protection by vaccination or by repeated infusions of bnAbs. In this study, we administered a recombinant bicistronic adeno-associated virus (AAV8) vector coding for both the light and heavy chains of the potent broadly neutralizing HIV-1 antibody VRC07 (AAV8-VRC07) to eight adults living with HIV. All participants remained on effective anti-retroviral therapy (viral load (VL) <50 copies per milliliter) throughout this phase 1, dose-escalation clinical trial ( NCT03374202 ). AAV8-VRC07 was given at doses of 5 × 1010, 5 × 1011 and 2.5 × 1012 vector genomes per kilogram by intramuscular (IM) injection. Primary endpoints of this study were to assess the safety and tolerability of AAV8-VRC07; to determine the pharmacokinetics and immunogenicity of in vivo VRC07 production; and to describe the immune response directed against AAV8-VRC07 vector and its products. Secondary endpoints were to assess the clinical effects of AAV8-VRC07 on CD4 T cell count and VL and to assess the persistence of VRC07 produced in participants. In this cohort, IM injection of AAV8-VRC07 was safe and well tolerated. No clinically significant change in CD4 T cell count or VL occurred during the 1-3 years of follow-up reported here. In participants who received AAV8-VRC07, concentrations of VRC07 were increased 6 weeks (P = 0.008) and 52 weeks (P = 0.016) after IM injection of the product. All eight individuals produced measurable amounts of serum VRC07, with maximal VRC07 concentrations >1 µg ml-1 in three individuals. In four individuals, VRC07 serum concentrations remained stable near maximal concentration for up to 3 years of follow-up. In exploratory analyses, neutralizing activity of in vivo produced VRC07 was similar to that of in vitro produced VRC07. Three of eight participants showed a non-idiotypic anti-drug antibody (ADA) response directed against the Fab portion of VRC07. This ADA response appeared to decrease the production of serum VRC07 in two of these three participants. These data represent a proof of concept that adeno-associated viral vectors can durably produce biologically active, difficult-to-induce bnAbs in vivo, which could add valuable new tools to the fight against infectious diseases.
Collapse
Affiliation(s)
- Joseph P Casazza
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Evan M Cale
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Galina V Yamshchikov
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Emily E Coates
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cynthia S Hendel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Laura Novik
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - LaSonji A Holman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alicia T Widge
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Preeti Apte
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ingelise Gordon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Martin R Gaudinski
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michelle Conan-Cibotti
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Martha C Nason
- Biostatistics Research Branch Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olga Trofymenko
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shinyi Telscher
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah H Plummer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Diane Wycuff
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - William C Adams
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Adrian McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Avery N Sukienik
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sijy O'Dell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason G Gall
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Britta Flach
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Travis L Terry
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Misook Choe
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wei Shi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Xuejun Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Florence Kaltovich
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Judy A Stein
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicole A Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard M Schwartz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Vaxart, Inc., South San Francisco, CA, USA
| | | | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Puhl DL, Mohanraj D, Nelson DW, Gilbert RJ. Designing electrospun fiber platforms for efficient delivery of genetic material and genome editing tools. Adv Drug Deliv Rev 2022; 183:114161. [PMID: 35183657 PMCID: PMC9724629 DOI: 10.1016/j.addr.2022.114161] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/29/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023]
Abstract
Electrospun fibers are versatile biomaterial platforms with great potential to support regeneration. Electrospun fiber characteristics such as fiber diameter, degree of alignment, rate of degradation, and surface chemistry enable the creation of unique, tunable scaffolds for various drug or gene delivery applications. The delivery of genetic material and genome editing tools via viral and non-viral vectors are approaches to control cellular protein production. However, immunogenicity, off-target effects, and low delivery efficiencies slow the progression of gene delivery strategies to clinical settings. The delivery of genetic material from electrospun fibers overcomes such limitations by allowing for localized, tunable delivery of genetic material. However, the process of electrospinning is harsh, and care must be taken to retain genetic material bioactivity. This review presents an up-to-date summary of strategies to incorporate genetic material onto or within electrospun fiber platforms to improve delivery efficiency and enhance the regenerative potential of electrospun fibers for various tissue engineering applications.
Collapse
Affiliation(s)
- Devan L Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, NY 12180, USA.
| | - Divya Mohanraj
- Department of Biological Sciences, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, NY 12180, USA.
| | - Derek W Nelson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, NY 12180, USA.
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, NY 12180, USA.
| |
Collapse
|
15
|
Dash S, Sharon DM, Mullick A, Kamen AA. ONLY A SMALL FRACTION OF CELLS PRODUCE ASSEMBLED CAPSIDS DURING TRANSFECTION-BASED MANUFACTURING OF ADENO-ASSOCIATED VIRUS VECTORS. Biotechnol Bioeng 2022; 119:1685-1690. [PMID: 35182435 PMCID: PMC9314941 DOI: 10.1002/bit.28068] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/14/2022] [Accepted: 02/13/2022] [Indexed: 11/14/2022]
Abstract
Plasmid transfection of mammalian cells is the dominant platform used to produce adeno‐associated virus (AAV) vectors for clinical and research applications. Low yields from this platform currently make it difficult to supply these activities with adequate material. In an effort to better understand the current limitations of transfection‐based manufacturing, this study examines what proportion of cells in a model transfection produce appreciable amounts of assembled AAV capsid. Using conformation‐specific antibody staining and flow cytometry, we report the surprising result that despite obtaining high transfection efficiencies and nominal vector yields in our model system, only 5%–10% of cells appear to produce measurable levels of assembled AAV capsids. This finding implies that considerable increases in vector titer could be realized through increasing the proportion of productive cells. Furthermore, we suggest that the flow cytometry assay used here to quantify productive cells may be a useful metric for future optimization of transfection‐based AAV vector manufacturing platforms.
Collapse
Affiliation(s)
- S Dash
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - D M Sharon
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - A Mullick
- Human Health Therapeutics, National Research Council of Canada, Montreal, QC, Canada
| | - A A Kamen
- Department of Bioengineering, McGill University, Montreal, QC, Canada.,Human Health Therapeutics, National Research Council of Canada, Montreal, QC, Canada
| |
Collapse
|
16
|
Chongchai A, Waramit S, Wongwichai T, Kampangtip J, Phitak T, Kongtawelert P, Hajitou A, Suwan K, Pothacharoen P. Targeting Human Osteoarthritic Chondrocytes with Ligand Directed Bacteriophage-Based Particles. Viruses 2021; 13:2343. [PMID: 34960616 PMCID: PMC8706358 DOI: 10.3390/v13122343] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/14/2021] [Accepted: 11/19/2021] [Indexed: 01/21/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by progressive deterioration and loss of articular cartilage. There is currently no treatment to reverse the onset of OA. Thus, we developed a targeted delivery strategy to transfer genes into primary human chondrocytes as a proof-of-concept study. We displayed a chondrocyte-affinity peptide (CAP) on the pIII minor coat protein of the M13 filamentous bacteriophage (phage)-based particle carrying a mammalian transgene cassette under cytomegalovirus CMV promoter and inverted terminal repeats (ITRs) cis elements of adeno-associated virus serotype 2 (AAV-2). Primary human articular chondrocytes (HACs) were used as an in vitro model, and the selectivity and binding properties of the CAP ligand in relation to the pathogenic conditions of HACs were characterized. We found that the CAP ligand is highly selective toward pathogenic HACs. Furthermore, the stability, cytotoxicity, and gene delivery efficacy of the CAP-displaying phage (CAP.Phage) were evaluated. We found that the phage particle is stable under a wide range of temperatures and pH values, while showing no cytotoxicity to HACs. Importantly, the CAP.Phage particle, carrying a secreted luciferase (Lucia) reporter gene, efficiently and selectively delivered transgene expression to HACs. In summary, it was found that the CAP ligand preferably binds to pathogenic chondrocytes, and the CAP.Phage particle successfully targets and delivers transgene to HACs.
Collapse
Affiliation(s)
- Aitthiphon Chongchai
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cancer Phagotherapy Group, Department of Brain Sciences, Burlington Danes Building, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Sajee Waramit
- Cancer Phagotherapy Group, Department of Brain Sciences, Burlington Danes Building, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Tunchanok Wongwichai
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jirawan Kampangtip
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thanyaluck Phitak
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Prachya Kongtawelert
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Amin Hajitou
- Cancer Phagotherapy Group, Department of Brain Sciences, Burlington Danes Building, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Keittisak Suwan
- Cancer Phagotherapy Group, Department of Brain Sciences, Burlington Danes Building, Hammersmith Hospital Campus, Imperial College London, London W12 0NN, UK
| | - Peraphan Pothacharoen
- Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
17
|
Abstract
The COVID-19 pandemic has motivated the rapid development of numerous vaccines that have proven effective against SARS-CoV-2. Several of these successful vaccines are based on the adenoviral vector platform. The mass manufacturing of these vaccines poses great challenges, especially in the context of a pandemic where extremely large quantities must be produced quickly at an affordable cost. In this work, two baseline processes for the production of a COVID-19 adenoviral vector vaccine, B1 and P1, were designed, simulated and economically evaluated with the aid of the software SuperPro Designer. B1 used a batch cell culture viral production step, with a viral titer of 5 × 1010 viral particles (VP)/mL in both stainless-steel and disposable equipment. P1 used a perfusion cell culture viral production step, with a viral titer of 1 × 1012 VP/mL in exclusively disposable equipment. Both processes were sized to produce 400 M/yr vaccine doses. P1 led to a smaller cost per dose than B1 ($0.15 vs. $0.23) and required a much smaller capital investment ($126 M vs. $299 M). The media and facility-dependent expenses were found to be the main contributors to the operating cost. The results indicate that adenoviral vector vaccines can be practically manufactured at large scale and low cost.
Collapse
|
18
|
Kim J, Vaughan HJ, Zamboni CG, Sunshine JC, Green JJ. High-throughput evaluation of polymeric nanoparticles for tissue-targeted gene expression using barcoded plasmid DNA. J Control Release 2021; 337:105-116. [PMID: 34097924 DOI: 10.1016/j.jconrel.2021.05.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/03/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022]
Abstract
Successful systemic gene delivery requires specific tissue targeting as well as efficient intracellular transfection. Increasingly, research laboratories are fabricating libraries of novel nanoparticles, engineering both new biomaterial structures and composition ratios of multicomponent systems. Yet, methods for screening gene delivery vehicles directly in vivo are often low-throughout, limiting the number of candidate nanoparticles that can be investigated. Here, we report a comprehensive, high-throughput method to evaluate a library of polymeric nanoparticles in vivo for tissue-specific gene delivery. The method involves pairing each nanoparticle formulation with a plasmid DNA (pDNA) that harbors a unique nucleotide sequence serving as the identifying "barcode". Using real time quantitative PCR (qPCR) for detection of the barcoded pDNA and quantitative reverse transcription PCR (RT-qPCR) for transcribed barcoded mRNA, we can quantify accumulation and transfection in tissues of interest. The barcode pDNA and primers were designed with sufficient sensitivity and specificity to evaluate multiple nanoparticle formulations per mouse, improving screening efficiency. Using this platform, we evaluated the biodistribution and transfection of 8 intravenously administered poly(beta-amino ester; PBAE) nanoparticle formulations, each with a PBAE polymer of differential structure. Significant levels of nanoparticle accumulation and gene transfection were observed mainly in organs involved in clearance, including spleen, liver, and kidneys. Interestingly, higher levels of transfection of select organs did not necessarily correlate with higher levels of tissue accumulation, highlighting the importance of directly measuring in vivo transfection efficiency as the key barcoded parameter in gene delivery vector optimization. To validate this method, nanoparticle formulations were used individually for luciferase pDNA delivery in vivo. The distribution of luciferase expression in tissues matched the transfection analysis by the barcode qPCR method, confirming that this platform can be used to accurately evaluate systemic gene delivery.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Hannah J Vaughan
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Camila G Zamboni
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Joel C Sunshine
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Departments of Neurosurgery, Oncology, and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Bisgin A, Sanlioglu AD, Eksi YE, Griffith TS, Sanlioglu S. Current Update on Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Development with a Special Emphasis on Gene Therapy Viral Vector Design and Construction for Vaccination. Hum Gene Ther 2021; 32:541-562. [PMID: 33858231 DOI: 10.1089/hum.2021.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome (SARS) is a newly emerging infectious disease (COVID-19) caused by the novel coronavirus SARS-coronavirus 2 (CoV-2). To combat the devastating spread of SARS-CoV-2, extraordinary efforts from numerous laboratories have focused on the development of effective and safe vaccines. Traditional live-attenuated or inactivated viral vaccines are not recommended for immunocompromised patients as the attenuated virus can still cause disease via phenotypic or genotypic reversion. Subunit vaccines require repeated dosing and adjuvant use to be effective, and DNA vaccines exhibit lower immune responses. mRNA vaccines can be highly unstable under physiological conditions. On the contrary, naturally antigenic viral vectors with well-characterized structure and safety profile serve as among the most effective gene carriers to provoke immune response via heterologous gene transfer. Viral vector-based vaccines induce both an effective cellular immune response and a humoral immune response owing to their natural adjuvant properties via transduction of immune cells. Consequently, viral vectored vaccines carrying the SARS-CoV-2 spike protein have recently been generated and successfully used to activate cytotoxic T cells and develop a neutralizing antibody response. Recent progress in SARS-CoV-2 vaccines, with an emphasis on gene therapy viral vector-based vaccine development, is discussed in this review.
Collapse
Affiliation(s)
- Atil Bisgin
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
- Department of Medical Genetics, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ahter D Sanlioglu
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Yunus Emre Eksi
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Thomas S Griffith
- The Department of Urology, School of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Salih Sanlioglu
- The Department of Gene and Cell Therapy, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
20
|
Characterization of Extracellular Vesicles Secreted in Lentiviral Producing HEK293SF Cell Cultures. Viruses 2021; 13:v13050797. [PMID: 33946875 PMCID: PMC8145507 DOI: 10.3390/v13050797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022] Open
Abstract
Lentiviral vectors (LVs) are a powerful tool for gene and cell therapy and human embryonic kidney cells (HEK293) have been extensively used as a platform for production of these vectors. Like most cells and cellular tissues, HEK293 cells release extracellular vesicles (EVs). EVs released by cells share similar size, biophysical characteristics and even a biogenesis pathway with cell-produced enveloped viruses, making it a challenge to efficiently separate EVs from LVs. Thus, EVs co-purified with LVs during downstream processing, are considered “impurities” in the context of gene and cell therapy. A greater understanding of EVs co-purifying with LVs is needed to enable improved downstream processing. To that end, EVs from an inducible lentivirus producing cell line were studied under two conditions: non-induced and induced. EVs were identified in both conditions, with their presence confirmed by transmission electron microscopy and Western blot. EV cargos from each condition were then further characterized by a multi-omic approach. Nineteen proteins were identified by mass spectrometry as potential EV markers to differentiate EVs in LV preparations. Lipid composition of EV preparations before and after LV induction showed similar enrichment in phosphatidylserine. RNA cargos in EVs showed enrichment in transcripts involved in viral processes and binding functions. These findings provide insights on the product profile of lentiviral preparations and could support the development of improved separation strategies aimed at removing co-produced EVs.
Collapse
|
21
|
Khan FA, Albalawi R, Pottoo FH. Trends in targeted delivery of nanomaterials in colon cancer diagnosis and treatment. Med Res Rev 2021; 42:227-258. [PMID: 33891325 DOI: 10.1002/med.21809] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022]
Abstract
Colon cancer is an adenocarcinoma, which subsequently develops into malignant tumors, if not treated properly. The current colon cancer therapy mainly revolves around chemotherapy, radiotherapy and surgery, but the search continues for more effective interventions. With the advancement of nanoparticles (NPs), it is now possible to diagnose and treat colon cancers with different types, shapes, and sizes of NPs. Nanoformulations such as quantum dots, iron oxide, polymeric NPs, dendrimers, polypeptides, gold NPs, silver NPs, platinum NPs, and cerium oxide have been either extensively used alone or in combination with other nanomaterials or drugs in colon cancer diagnosis, and treatments. These nanoformulations possess high biocompatibility and bioavailability, which makes them the most suitable candidates for cancer treatment. The size and shape of NPs are critical to achieving an effective drug delivery in cancer treatment and diagnosis. Most NPs currently are under different testing phases (in vitro, preclinical, and clinical), whereas some of them have been approved for therapeutic applications. We have comprehensively reviewed the recent advances in the applications of NPs-based formulations in colon cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Firdos A Khan
- Department of Stem Cell Biology, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Reem Albalawi
- Department of Stem Cell Biology, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Student of the volunteer/training program at IRMC
| | - Faheem H Pottoo
- College of Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
22
|
Kawka K, Wilton AN, Madadkar P, Medina MFC, Lichty BD, Ghosh R, Latulippe DR. Integrated development of enzymatic DNA digestion and membrane chromatography processes for the purification of therapeutic adenoviruses. Sep Purif Technol 2021. [DOI: 10.1016/j.seppur.2020.117503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
23
|
Petrich J, Marchese D, Jenkins C, Storey M, Blind J. Gene Replacement Therapy: A Primer for the Health-system Pharmacist. J Pharm Pract 2020; 33:846-855. [PMID: 31248331 PMCID: PMC7675776 DOI: 10.1177/0897190019854962] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Comprehensive review of gene replacement therapy with guidance and expert opinion on handling and administration for pharmacists. SUMMARY There are currently ∼2600 gene therapy clinical trials worldwide and 4 Food and Drug Administration (FDA)-approved gene therapy products available in the United States. Gene therapy and its handling are different from other drugs; however, there is a lack of guidance from the National Institutes of Health (NIH), FDA, Centers for Disease Control and Prevention (CDC), World Health Organization (WHO), and professional associations regarding their pharmaceutical application. Although the NIH stratifies the backbone biologicals of viral vectors in gene therapies into risk groups, incomplete information regarding minimization of exposure and reduction of risk exists. In the absence of defined guidance, individual institutions develop their own policies and procedures, which often differ and are often outdated. This review provides expert opinion on the role of pharmacists in institutional preparedness, as well as gene therapy handling and administration. A suggested infrastructural model for gene replacement therapy handling is described, including requisite equipment acquisition and standard operating procedure development. Personnel, patient, and caregiver education and training are discussed. CONCLUSION Pharmacists have a key role in the proper handling and general management of gene replacement therapies, identifying risk level, establishing infrastructure, and developing adequate policies and protocols, particularly in the absence of consensus guidelines for the handling and transport of gene replacement therapies.
Collapse
Affiliation(s)
- John Petrich
- Department of Pharmacy, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | - Chris Jenkins
- Clinical Biosafety Services, LLC, St. Louis, MO, USA
| | - Michael Storey
- Department of Pharmacy, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Jill Blind
- Department of Pharmacy, Nationwide Children’s Hospital, Columbus, OH, USA
| |
Collapse
|
24
|
Emerson J, Kara B, Glassey J. Multivariate data analysis in cell gene therapy manufacturing. Biotechnol Adv 2020; 45:107637. [PMID: 32980438 DOI: 10.1016/j.biotechadv.2020.107637] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/27/2020] [Accepted: 09/22/2020] [Indexed: 01/26/2023]
Abstract
The emergence of cell gene therapy (CGT) as a safe and efficacious treatment for numerous severe inherited and acquired human diseases has led to growing interest and investment in new CGT products. The most successful of these have been autologous viral vector-based treatments. The development of viral vector manufacturing processes and ex vivo patient cell processing capabilities is a pressing issue in the advancement of autologous viral vector-based CGT treatments. In viral vector production, scale-up is a critical task due to the limited scalability of traditional laboratory systems and the demand for high volumes of viral vector manufactured in accordance with current good manufacturing practice. Ex vivo cell processing methods require optimisation and automation before they can be scaled out, and several other manufacturing challenges are prevalent such as high levels of raw material and process variability, difficulty characterising complex materials, and a lack of knowledge of critical process parameters and their effect on critical quality attributes of the viral vector and cell drug products. Multivariate data analysis (MVDA) has been leveraged successfully in a variety of applications in the chemical and biochemical industries, including for tasks such as bioprocess monitoring, identification of critical process parameters and assessment of process variability and comparability during process development, scale-up and technology transfer. Henceforth, MVDA is reviewed here as a suitable tool for tackling some of the challenges faced in the development of CGT manufacturing processes. A summary of some key CGT manufacturing challenges is provided along with a review of MVDA applications to mammalian and microbial processes, and an exploration of the potential benefits, requirements and pre-requisites of MVDA applications in the development of CGT manufacturing processes.
Collapse
Affiliation(s)
- Joseph Emerson
- School of Engineering, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| | - Bo Kara
- Currently, Evox Therapeutics, Medawar Centre, Oxford OX4 4HG, UK.
| | - Jarka Glassey
- School of Engineering, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
25
|
Cheng L, Ma D, Lu L, Ouyang D, Xi Z. Building Customizable Multisite‐Targeting c‐Myc shRNA Array into Branch‐PCR‐Constructed DNA Nanovectors for Enhanced Tumor Cell Suppression. ChemistrySelect 2020. [DOI: 10.1002/slct.202002609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Longhuai Cheng
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| | - Dejun Ma
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| | - Liqing Lu
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| | - Di Ouyang
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| | - Zhen Xi
- Department of Chemical Biology State Key Laboratory of Elemento-Organic Chemistry National Engineering Research Center of Pesticide (Tianjin) College of Chemistry Nankai University Weijin Road 94 Tianjin 300071 P. R. China
| |
Collapse
|
26
|
Enhanced Production of Herpes Simplex Virus 1 Amplicon Vectors by Gene Modification and Optimization of Packaging Cell Growth Medium. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:491-496. [PMID: 32258212 PMCID: PMC7114837 DOI: 10.1016/j.omtm.2020.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/10/2020] [Indexed: 01/25/2023]
Abstract
Herpes simplex virus 1 (HSV-1)-derived amplicon vectors are unique in their ability to accommodate large DNA molecules allowing whole genomic loci to be included with all of their regulatory elements. Additional advantages of these amplicons include their minimal toxicity and ability to persist as episomes, with negligible risk of insertional mutagenesis, being particularly well-suited for gene therapy of neurological disorders due to their outstanding ability to deliver genes into neurons and other neural cells. However, extensive gene therapy application has been hindered by difficulties in vector production. This work improved HSV-1 amplicons production by genetic modification of the packaging cell line and optimization of the culture medium. A stably-transfected Vero 2-2 cell line overexpressing the anti-apoptotic Bcl-2 protein was generated, exhibiting an increased resistance to apoptosis, prolonged culture duration, and a significant improvement in viral vector production. Additionally, supplementation of the growth medium with antioxidants, polyamines, amino acids, and reduced glutathione further increased the yield of packaged amplicon vectors. With these modifications, HSV-1 amplicons could be isolated from culture supernatants instead of cell lysates, leading to vector preparations with higher titer and purity and paving the way for generation of stable cell lines that are capable of continuous herpesviral vector production.
Collapse
|
27
|
Do Minh A, Tran MY, Kamen AA. Lentiviral Vector Production in Suspension Culture Using Serum-Free Medium for the Transduction of CAR-T Cells. Methods Mol Biol 2020; 2086:77-83. [PMID: 31707669 DOI: 10.1007/978-1-0716-0146-4_6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The production of lentiviral vectors (LVs) in human embryonic kidney 293 (HEK293) cells using serum-free medium in a suspension culture for the transduction of chimeric antigen receptor T-cells (CAR-T) can be achieved by different methods. This chapter describes LV production by transient transfection, induction of stable packaging cell lines, and induction of stable producer cell lines.
Collapse
Affiliation(s)
- Aline Do Minh
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Michelle Yen Tran
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Amine A Kamen
- Department of Bioengineering, McGill University, Montreal, QC, Canada.
| |
Collapse
|
28
|
Applications of Spherical Nucleic Acid Nanoparticles as Delivery Systems. Trends Mol Med 2019; 25:1066-1079. [DOI: 10.1016/j.molmed.2019.08.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
|
29
|
Wang F, Qin Z, Lu H, He S, Luo J, Jin C, Song X. Clinical translation of gene medicine. J Gene Med 2019; 21:e3108. [PMID: 31246328 DOI: 10.1002/jgm.3108] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/30/2019] [Accepted: 06/13/2019] [Indexed: 02/05/2023] Open
Abstract
Gene therapy has recently witnessed accelerated progress as a new therapeutic strategy with the potential to treat a range of inherited and acquired diseases. Billions of dollars have been invested in basic and clinical research on gene medicine, with ongoing clinical trials focused on cancer, monogenic diseases, cardiovascular diseases and other refractory diseases. Advances addressing the inherent challenges of gene therapy, particularly those related to retaining the delivery efficacy and minimizing unwanted immune responses, provide the basis for the widespread clinical application of gene medicine. Several types of genes delivered by viral or non-viral delivery vectors have demonstrated encouraging results in both animals and humans. As augmented by clinical indications, gene medicine techniques have rapidly become a promising alternative to conventional therapeutic strategies because of their better clinical benefit and lower toxicities. Their application in the clinic has been extensive as a result of the approval of many gene therapy drugs in recent years. In this review, we provide a comprehensive overview of the clinical translation of gene medicine, focusing on the key events and latest progress made regarding clinical gene therapy products. We also discuss the gene types and non-viral materials with respect to developing gene therapeutics in clinical trials.
Collapse
Affiliation(s)
- Fazhan Wang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Zhou Qin
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Hansi Lu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Siyan He
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jing Luo
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Chaohui Jin
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiangrong Song
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
MicroRNA-34 family: a potential tumor suppressor and therapeutic candidate in cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:53. [PMID: 30717802 PMCID: PMC6360685 DOI: 10.1186/s13046-019-1059-5] [Citation(s) in RCA: 331] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/27/2019] [Indexed: 12/20/2022]
Abstract
MicroRNA-34 (miR-34) has been reported to be dysregulated in various human cancers and regarded as a tumor suppressive microRNA because of its synergistic effect with the well-known tumor suppressor p53. Along with the application of MRX34, the first tumor-targeted microRNA drug which based on miR-34a mimics, on phase I clinical trial (NCT01829971), the significance of miR-34 is increasingly recognized. miR-34 plays a crucial role on repressing tumor progression by involving in epithelial-mesenchymal transition (EMT) via EMT- transcription factors, p53 and some important signal pathways. Not only that, numerous preclinical researches revealed the giant potential of miR-34a on cancer therapy through diversiform nano-scaled delivery systems. Here, we provide an overview about the function of miR-34 in various cancers and the mechanism of miR-34 in tumor-associated EMT. Furthermore, its potential role as a microRNA therapeutic candidate is also discussed. Notwithstanding some obstacles existed, the extensive application prospect of miR-34 on oncotherapy cannot be neglected.
Collapse
|
31
|
Xie L, Miao J, Li X, Yi X, Chu J. Regulation of the pyruvate metabolism node by monogene and polygene engineering of HEK-293 cells. RSC Adv 2019; 9:35760-35770. [PMID: 35528064 PMCID: PMC9074685 DOI: 10.1039/c9ra07418j] [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: 09/15/2019] [Accepted: 10/21/2019] [Indexed: 11/21/2022] Open
Abstract
HEK-293 cells are increasingly being used in the production of human adenovirus (HAdV) vaccines. However, the production of HAdV vaccine has not met the requirements of industrial production. Recently, we investigated the effects of various regulatory genes of the pyruvate metabolism node on the substance and energy metabolism and adenovirus reproduction in HEK-293 cells. Initially, single regulatory genes, including pkm2, pdhα, pyc2, mpc3, aralar1, ldha and pdk1, were studied. We found that metabolic performance and adenovirus reproduction capacity in HEK-293 cells were improved, and maximum adenovirus titre was increased approximately 15-fold. Next, we co-overexpressed the key genes, including pkm2, pyc2 and aralar1. The PYC2-A-P-L cells that had the appropriate co-overexpression levels of three genes had the most pronounced regulatory effect. The maximum cell density and maximum specific growth rate were increased by 21% compared with that in the control. The ΔLac/ΔGlc and ΔNH3/ΔGln were decreased by 26% and 27%, respectively. The ATP production rate and the ATP/O2 ratio were increased by 110% and 20%, respectively. The level of reactive oxygen species (ROS) was reduced by 60%. The adenovirus reproductive ability of the PYC2-A-P-L cells was approximately 30-fold higher than that of the control. The results showed that proper overexpression of the aralar1, pkm2 and pyc2 genes can significantly improve the substance and energy metabolism efficiency in HEK-293 cells, maximize the metabolic balance of pyruvate, and ultimately improve HAdV reproduction. This study provides a method of regulation of pyruvate metabolism and polygenic metabolic engineering in mammalian cells cultured in vitro and suggests an effective method for efficient HAdV production. HEK-293 cells are increasingly being used in the production of human adenovirus (HAdV) vaccines.![]()
Collapse
Affiliation(s)
- Li Xie
- State Key Laboratory of Bioreactor Engineering
- School of Bioengineering, East China University of Science and Technology
- Shanghai 200237
- China
| | - Junqing Miao
- State Key Laboratory of Bioreactor Engineering
- School of Bioengineering, East China University of Science and Technology
- Shanghai 200237
- China
| | - Xiangchao Li
- State Key Laboratory of Bioreactor Engineering
- School of Bioengineering, East China University of Science and Technology
- Shanghai 200237
- China
| | - Xiaoping Yi
- State Key Laboratory of Bioreactor Engineering
- School of Bioengineering, East China University of Science and Technology
- Shanghai 200237
- China
| | - Ju Chu
- State Key Laboratory of Bioreactor Engineering
- School of Bioengineering, East China University of Science and Technology
- Shanghai 200237
- China
| |
Collapse
|
32
|
Stuible M, van Lier F, Croughan MS, Durocher Y. Beyond preclinical research: production of CHO-derived biotherapeutics for toxicology and early-phase trials by transient gene expression or stable pools. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
33
|
Zhang ZK, Li J, Guan D, Liang C, Zhuo Z, Liu J, Lu A, Zhang G, Zhang BT. Long Noncoding RNA lncMUMA Reverses Established Skeletal Muscle Atrophy following Mechanical Unloading. Mol Ther 2018; 26:2669-2680. [PMID: 30415659 PMCID: PMC6225098 DOI: 10.1016/j.ymthe.2018.09.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/13/2018] [Accepted: 09/14/2018] [Indexed: 12/26/2022] Open
Abstract
Reversing established muscle atrophy following mechanical unloading is of great clinical challenge. Long noncoding RNAs (lncRNAs) have been demonstrated to play important roles in myogenesis. Here we identified a lncRNA (mechanical unloading-induced muscle atrophy-related lncRNA [lncMUMA]) enriched in muscle, which was the most downregulated lncRNA during muscle atrophy development in hindlimb suspension (HLS) mice. The in vitro and in vivo data demonstrated that the decreased expression levels of lncMUMA closely associated with a reduction of myogenesis during mechanical unloading. Mechanistically, lncMUMA promoted myogenic differentiation by functioning as a miR-762 sponge to regulate the core myogenic regulator MyoD in vitro. The enforced expression of lncMUMA relieved the decreases in MyoD protein and muscle mass in miR-762 knockin mice. Therapeutically, the enforced expression of lncMUMA improved the in vitro myogenic differentiation of myoblasts under microgravity simulation, prevented the muscle atrophy development, and reversed the established muscle atrophy in HLS mice. These findings identify lncMUMA as an anabolic regulator to reverse established muscle atrophy following mechanical unloading.
Collapse
Affiliation(s)
- Zong-Kang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Jie Li
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Daogang Guan
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
| | - Chao Liang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
| | - Zhenjian Zhuo
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Jin Liu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
| | - Aiping Lu
- Institute of Integrated Bioinformedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China.
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China.
| |
Collapse
|
34
|
Deng YW, Hao WJ, Li YW, Li YX, Zhao BC, Lu D. Hsa-miRNA-143-3p Reverses Multidrug Resistance of Triple-Negative Breast Cancer by Inhibiting the Expression of Its Target Protein Cytokine-Induced Apoptosis Inhibitor 1 In Vivo. J Breast Cancer 2018; 21:251-258. [PMID: 30275853 PMCID: PMC6158160 DOI: 10.4048/jbc.2018.21.e40] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/17/2018] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Multidrug resistance (MDR) remains a major obstacle in the treatment of triple-negative breast cancer (TNBC) with conventional chemotherapeutic agents. A previous study demonstrated that hsa-miRNA-143-3p plays a vital role in drug resistance of TNBC. Downregulation of hsa-miRNA-143-3p upregulated the expression of its target protein cytokine-induced apoptosis inhibitor 1 (CIAPIN1) in order to activate MDR, while upregulation of hsa-miRNA-143-3p effectively enhances the sensitivity of drug-resistant TNBC cells to chemotherapeutics. The present study aimed to further verify these findings in vivo. METHODS We established a hypodermic tumor nude mice model using paclitaxel-resistant TNBC cells. We expressed ectopic hsa-miRNA-143-3p under the control of a breast cancer-specific human mammaglobin promoter that guided the efficient expression of exogenous hsa-miRNA-143-3p only in breast cancer cells. Thereafter, we overexpressed hsa-miRNA-143-3p in xenografts using a recombinant virus system and quantified the expression of hsa-miRNA-143-3p, CIAPIN1 protein, and proteins encoded by related functional genes by western blot. RESULTS We successfully completed the prospective exploration of the intravenous virus injection pattern from extensive expression to targeted expression. The overexpression of hsa-miRNA-143-3p significantly alleviated chemoresistance of TNBC by inhibiting viability. In addition, we observed that the expression of CIAPIN1 as a hsa-miRNA-143-3p target protein was remarkably decreased. CONCLUSION We partly illustrated the mechanism underlying the hsa-miRNA-143-3p/CIAPIN1 drug resistance pathway. HsamiRNA-143-3p as a tumor suppressive microRNA may be a novel target to effectively reverse MDR of TNBC in vivo.
Collapse
Affiliation(s)
- Yu Wei Deng
- Department of Medical Oncology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wen Jing Hao
- Department of Medical Oncology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Wen Li
- Department of Medical Oncology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Xin Li
- Department of Medical Oncology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Chen Zhao
- Department of Medical Oncology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dan Lu
- Department of Medical Oncology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
35
|
Chen KD, Wu XX, Yu DS, Ou HL, Li YH, Zhou YQ, Li LJ. Process optimization for the rapid production of adenoviral vectors for clinical trials in a disposable bioreactor system. Appl Microbiol Biotechnol 2018; 102:6469-6477. [PMID: 29858958 DOI: 10.1007/s00253-018-9091-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 11/28/2022]
Abstract
Recombinant adenoviral (Ad) vectors are highly efficient gene transfer vectors widely used in vaccine development and immunotherapy. To promote the industrial application of Ad vectors, studies focusing on reducing the cost of manufacturing, shortening the preclinical research period, and improving the quality of products are needed. Here, we describe a highly efficient and economical process for producing Ad vector in a novel, single-use bioreactor system suitable for clinical trials. A mini-bioreactor was used for parameter optimization and development of medium replacement protocols for Ad5-GFP production before scale-up. HEK293 cell culture and virus infection were monitored in a disposable AmProtein Current Perfusion Bioreactor and Bioflo310 bioreactor using optimized parameters and medium replacement protocols. The total cell number increased from 2.0 × 109 to 3.2 × 1010 after 6 days of culture. The total number of viral particles obtained in a single batch was 1.2 × 1015. These results demonstrate the efficiency and suitability of this system for Ad vector production for research and GMP applications.
Collapse
Affiliation(s)
- Ke-Da Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovative Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiao-Xin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovative Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Dong-Shan Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovative Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Hui-Lin Ou
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovative Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yan-Hua Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovative Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yu-Qing Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovative Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lan-Juan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovative Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
36
|
Li J, Liang H, Liu J, Wang Z. Poly (amidoamine) (PAMAM) dendrimer mediated delivery of drug and pDNA/siRNA for cancer therapy. Int J Pharm 2018; 546:215-225. [PMID: 29787895 DOI: 10.1016/j.ijpharm.2018.05.045] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022]
Abstract
Poly (amidoamine) (PAMAM) dendrimers are well-defined, highly branched macromolecules with numerous active amine groups on the surface. Because of their unique properties, PAMAM dendrimers have steadily grown in popularity in drug delivery, gene therapy, medical imaging and diagnostic application. This review focuses on the recent developments on the application in PAMAM dendrimers as effective carriers for drug and gene (pDNA, siRNA) delivery in cancer therapy, including: a) PAMAM for anticancer drug delivery; b) PAMAM and gene therapy; c) PAMAM used in overcoming tumor multidrug resistance; d) PAMAM used for hybrid nanoparticles; and e) PAMAM linked or loaded in other nanoparticles.
Collapse
Affiliation(s)
- Jun Li
- School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.
| | - Huamin Liang
- Institute of Technology Innovation, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230088, Anhui, China
| | - Jing Liu
- Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing 100084, China
| | - Ziyuan Wang
- School of Life Sciences, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| |
Collapse
|
37
|
Pierson CR. Gene therapy strategies for X-linked myotubular myopathy. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1443807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Christopher R. Pierson
- Department of Pathology and Laboratory Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
- Departments of Pathology and Biomedical Education & Anatomy, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
38
|
The Role of Natural-Based Biomaterials in Advanced Therapies for Autoimmune Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:127-146. [DOI: 10.1007/978-981-13-0947-2_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|