Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Processes ; 11(3), 2023.
Article in English | Scopus | ID: covidwho-2296375

ABSTRACT

The production of messenger ribonucleic acid (mRNA) and other biologics is performed primarily in batch mode. This results in larger equipment, cleaning/sterilization volumes, and dead times compared to any continuous approach. Consequently, production throughput is lower and capital costs are relatively high. Switching to continuous production thus reduces the production footprint and also lowers the cost of goods (COG). During process development, from the provision of clinical trial samples to the production plant, different plant sizes are usually required, operating at different operating parameters. To speed up this step, it would be optimal if only one plant with the same equipment and piping could be used for all sizes. In this study, an efficient solution to this old challenge in biologics manufacturing is demonstrated, namely the qualification and validation of a plant setup for clinical trial doses of about 1000 doses and a production scale-up of about 10 million doses. Using the current example of the Comirnaty BNT162b2 mRNA vaccine, the cost-intensive in vitro transcription was first optimized in batch so that a yield of 12 g/L mRNA was achieved, and then successfully transferred to continuous production in the segmented plug flow reactor with subsequent purification using ultra- and diafiltration, which enables the recycling of costly reactants. To realize automated process control as well as real-time product release, the use of appropriate process analytical technology is essential. This will also be used to efficiently capture the product slug so that no product loss occurs and contamination from the fill-up phase is <1%. Further work will focus on real-time release testing during a continuous operating campaign under autonomous operational control. Such efforts will enable direct industrialization in collaboration with appropriate industry partners, their regulatory affairs, and quality assurance. A production scale-operation could be directly supported and managed by data-driven decisions. © 2023 by the authors.

2.
Biotechnol Bioeng ; 2022 Dec 05.
Article in English | MEDLINE | ID: covidwho-2244351

ABSTRACT

The COVID-19 pandemic triggered an unprecedented rate of development of mRNA vaccines, which are produced by in vitro transcription reactions. The latter has been the focus of intense development to increase productivity and decrease cost. Optimization of IVT depends on understanding of the impact of individual reagents on the kinetics of mRNA production and the consumption of building blocks, which is hampered by slow, low-throughput, end-point analytics. We implemented a workflow based on rapid at-line HPLC monitoring of consumption of NTPs with concomitant production of mRNA, with a sub-3 min read-out, allowing for adjustment of IVT reaction parameters with minimal lag. IVT was converted to fed-batch resulting in doubling the reaction yield compared to batch IVT protocol, reaching 10 mg/mL for multiple constructs. When coupled with exonuclease digestion, HPLC analytics for quantification of mRNA was extended to monitoring capping efficiency of produced mRNA. When HPLC monitoring was applied to production of an ARCA-capped mRNA construct, which requires an approximate 4:1 ARCA:GTP ratio, the optimized fed-batch approach achieved productivity of 9 mg/mL with 79% capping. The study provides a methodological platform for optimization of factors influencing IVT reactions, converting the reaction from batch to fed-batch mode, determining reaction kinetics, which are critical for optimization of continuous addition of reagents, thereby paving the way towards continuous manufacturing of mRNA. This article is protected by copyright. All rights reserved.

3.
RNA Technologies ; 13:41-71, 2022.
Article in English | Scopus | ID: covidwho-2059695

ABSTRACT

Last decade has witnessed tremendous growth in the new promising treatment options based on mRNA, RNAi, antisense RNA, and RNA aptamers, the four classes of RNA-based therapeutics. Among these, mRNA-based therapy is centered on producing proteins within the cells to supplant deficient or abnormal proteins and in vaccination to a target pathogen. The potential of mRNA therapeutics is evident from the two major mRNA vaccines approved for COVID-19: developed by Moderna and by Pfizer. Nonetheless, mRNA therapeutic potential extends far beyond this, such as in treating genetic diseases, cancers, and other infectious diseases. Given the potential of mRNA therapeutics, this chapter is written to provide the reader an insight into the features of several synthetic mRNA platforms, production, purification;strategies to increase the stability and reduce the immunogenicity of therapeutic mRNA molecules;delivery methods of these mRNAs in vivo;and their applications, safety, and efficacy. Graphical : A detailed diagram of the chemically modified mRNA, with the in vitro delivery modes on the left, and the myriad of applications, namely the treatment of major genetic diseases on the right. The IVT mRNA is represented with more details above the diagram. © 2022, The Author(s), under exclusive license to Springer Nature Switzerland AG.

4.
Chemie Ingenieur Technik (CIT) ; : 1, 2022.
Article in English | Academic Search Complete | ID: covidwho-2059345

ABSTRACT

The COVID‐19 pandemic triggered an unprecedented surge in development of mRNA‐based vaccines. Despite the need to increase process productivity and thus decrease the cost of mRNA vaccines, limited scientific literature is available on strategies to increase the yield of in vitro transcription (IVT) reaction, the unit operation with highest cost of goods, which has traditionally been performed as a batch reaction. Single‐use bioreactors are traditionally used for cell‐based production of biopharmaceuticals, but some core functionalities, such as controlled and automated feed addition, are potentially useful for cell‐free mRNA processes. We report the production of 2 g mRNA in an Ambr® 250 Modular bioreactor system with a starting volume of 100 mL, reaching a maximum mRNA concentration of 12 g L−1 by a fed‐batch IVT approach, and demonstrate the feasibility of continuous fed‐batch production, paving the way towards continuous manufacturing of mRNA. [ FROM AUTHOR] Copyright of Chemie Ingenieur Technik (CIT) is the property of John Wiley & Sons, Inc. and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full . (Copyright applies to all s.)

5.
RNA Therapeutics: The Evolving Landscape of RNA Therapeutics ; : 141-169, 2022.
Article in English | Scopus | ID: covidwho-2027784

ABSTRACT

RNA molecules are prone to nuclease degradation and recognition by the innate immune system. Chemical modifications of the phosphate backbone, sugar, and/or nucleobase have helped increase resistance to degradation, while reducing recognition by immune sensors, and have proven crucial for the bench-to-bedside translation of several small RNA-based therapeutics (i.e., Onpattro and Givlaari). RNA molecules produced using in vitro transcription (IVT) demonstrated superior performance compared to other vaccine platforms gaining prominence in the fight against the COVID-19 pandemic (i.e., Comirnaty and Spikevax). This chapter discusses the elements of RNA recognition by innate immune sensors, the origins of the immunogenicity of in vitro transcribed RNA, as well as strategies to mitigate immunogenicity and improve translation of IVT-produced mRNA. We further discuss different nucleoside modifications and their influence on the capacity of RNA to activate the innate immune system and improve the therapeutic potential of mRNA. © 2022 Elsevier Inc. All rights reserved.

6.
Methods Mol Biol ; 2573: 77-87, 2022.
Article in English | MEDLINE | ID: covidwho-2007118

ABSTRACT

Modified mRNA (modRNA) is a safe and effective vector for gene-based therapies. Notably, the safety of modRNA has been validated through COVID-19 vaccines which incorporate modRNA technology to translate spike proteins. Alternative gene delivery methods using plasmids, lentiviruses, adenoviruses, and adeno-associated viruses have suffered from key challenges such as genome integration, delayed and uncontrolled expression, and immunogenic responses. However, modRNA poses no risk of genome integration, has transient and rapid expression, and lacks an immunogenic response. Our lab utilizes modRNA-based therapies to promote cardiac regeneration following myocardial infarction and heart failure. We have also developed and refined an optimized and economical method for synthesis of modRNA. Here, we provide an updated methodology with improved translational efficiency for in vitro and in vivo application.


Subject(s)
COVID-19 Vaccines , COVID-19 , COVID-19/therapy , Gene Transfer Techniques , Genetic Therapy/methods , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Int Rev Cell Mol Biol ; 372: 295-316, 2022.
Article in English | MEDLINE | ID: covidwho-1894712

ABSTRACT

mRNA vaccines have been increasingly recognized as a powerful vaccine platform since the FDA approval of two COVID-19 mRNA vaccines, which demonstrated outstanding prevention efficacy as well as great safety profile. Notably, nucleoside modification and lipid nanoparticle-facilitated delivery has greatly improved the immunogenicity, stability, and translation efficiency of mRNA molecule. Here we review the recent progress in mRNA vaccine development, including nucleoside modification, in vitro synthesis and product purification, and lipid nanoparticle vectors for in vivo delivery and efficient translation. We also briefly introduce the clinical application of mRNA vaccine in preventing infectious diseases and treating inflammatory diseases including cancer.


Subject(s)
COVID-19 , Nanoparticles , COVID-19/prevention & control , Humans , Liposomes , Nucleosides , RNA, Messenger/genetics , Vaccines, Synthetic , mRNA Vaccines
8.
Mol Pharm ; 19(4): 1047-1058, 2022 04 04.
Article in English | MEDLINE | ID: covidwho-1721386

ABSTRACT

The coronavirus disease of 2019 (COVID-19) pandemic launched an unprecedented global effort to rapidly develop vaccines to stem the spread of the novel severe acute respiratory syndrome coronavirus (SARS-CoV-2). Messenger ribonucleic acid (mRNA) vaccines were developed quickly by companies that were actively developing mRNA therapeutics and vaccines for other indications, leading to two mRNA vaccines being not only the first SARS-CoV-2 vaccines to be approved for emergency use but also the first mRNA drugs to gain emergency use authorization and to eventually gain full approval. This was possible partly because mRNA sequences can be altered to encode nearly any protein without significantly altering its chemical properties, allowing the drug substance to be a modular component of the drug product. Lipid nanoparticle (LNP) technology required to protect the ribonucleic acid (RNA) and mediate delivery into the cytoplasm of cells is likewise modular, as are technologies and infrastructure required to encapsulate the RNA into the LNP. This enabled the rapid adaptation of the technology to a new target. Upon the coattails of the clinical success of mRNA vaccines, this modularity will pave the way for future RNA medicines for cancer, gene therapy, and RNA engineered cell therapies. In this review, trends in the publication records and clinical trial registrations are tallied to show the sharp intensification in preclinical and clinical research for RNA medicines. Demand for the manufacturing of both the RNA drug substance (DS) and the LNP drug product (DP) has already been strained, causing shortages of the vaccine, and the rise in development and translation of other mRNA drugs in the coming years will exacerbate this strain. To estimate demand for DP manufacturing, the dosing requirements for the preclinical and clinical studies of the two approved mRNA vaccines were examined. To understand the current state of mRNA-LNP production, current methods and technologies are reviewed, as are current and announced global capacities for commercial manufacturing. Finally, a vision is rationalized for how emerging technologies such as self-amplifying mRNA, microfluidic production, and trends toward integrated and distributed manufacturing will shape the future of RNA manufacturing and unlock the potential for an RNA medicine revolution.


Subject(s)
COVID-19 , COVID-19 Vaccines , Humans , Liposomes , Nanoparticles , RNA, Messenger/metabolism , SARS-CoV-2/genetics
9.
Mol Ther Nucleic Acids ; 27: 854-869, 2022 Mar 08.
Article in English | MEDLINE | ID: covidwho-1615681

ABSTRACT

In vitro transcribed (IVT)-mRNA has been accepted as a promising therapeutic modality. Advances in facile and rapid production technologies make IVT-mRNA an appealing alternative to protein- or virus-based medicines. Robust expression levels, lack of genotoxicity, and their manageable immunogenicity benefit its clinical applicability. We postulated that innate immune responses of therapeutically relevant human cells can be tailored or abrogated by combinations of 5'-end and internal IVT-mRNA modifications. Using primary human macrophages as targets, our data show the particular importance of uridine modifications for IVT-mRNA performance. Among five nucleotide modification schemes tested, 5-methoxy-uridine outperformed other modifications up to 4-fold increased transgene expression, triggering moderate proinflammatory and non-detectable antiviral responses. Macrophage responses against IVT-mRNAs exhibiting high immunogenicity (e.g., pseudouridine) could be minimized upon HPLC purification. Conversely, 5'-end modifications had only modest effects on mRNA expression and immune responses. Our results revealed how the uptake of chemically modified IVT-mRNA impacts human macrophages, responding with distinct patterns of innate immune responses concomitant with increased transient transgene expression. We anticipate our findings are instrumental to predictively address specific cell responses required for a wide range of therapeutic applications from eliciting controlled immunogenicity in mRNA vaccines to, e.g., completely abrogating cell activation in protein replacement therapies.

10.
Biomedicines ; 9(5)2021 May 10.
Article in English | MEDLINE | ID: covidwho-1389281

ABSTRACT

Lipid nanoparticle (LNP) formulated messenger RNA-based (LNP-mRNA) vaccines came into the spotlight as the first vaccines against SARS-CoV-2 virus to be applied worldwide. Long-known benefits of mRNA-based technologies consisting of relatively simple and fast engineering of mRNA encoding for antigens and proteins of interest, no genomic integration, and fast and efficient manufacturing process compared with other biologics have been verified, thus establishing a basis for a broad range of applications. The intrinsic immunogenicity of LNP formulated in vitro transcribed (IVT) mRNA is beneficial to the LNP-mRNA vaccines. However, avoiding immune activation is critical for therapeutic applications of LNP-mRNA for protein replacement where targeted mRNA expression and repetitive administration of high doses for a lifetime are required. This review summarizes our current understanding of immune activation induced by mRNA, IVT byproducts, and LNP. It gives a comprehensive overview of the present status of preclinical and clinical studies in which LNP-mRNA is used for protein replacement and treatment of rare diseases with an emphasis on safety. Moreover, the review outlines innovations and strategies to advance pharmacology and safety of LNP-mRNA for non-immunotherapy applications.

11.
Emerg Microbes Infect ; 10(1): 1217-1218, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1276098

ABSTRACT

A simple mRNA vaccine was shown to protect mice against tuberculosis more than 15 years ago. Like COVID-19, tuberculosis is a respiratory infection killing over a million people per year. It too presents a global emergency. Can the stunning success of RNA vaccination against COVID-19 be replicated for TB?


Subject(s)
Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Vaccines, Synthetic/immunology , Humans , Tuberculosis/immunology , Tuberculosis/microbiology
12.
Curr Protoc ; 1(2): e39, 2021 Feb.
Article in English | MEDLINE | ID: covidwho-1064340

ABSTRACT

Synthetic messenger RNA (mRNA)-based therapeutics are an increasingly popular approach to gene and cell therapies, genome engineering, enzyme replacement therapy, and now, during the global SARS-CoV-2 pandemic, vaccine development. mRNA for such purposes can be synthesized through an enzymatic in vitro transcription (IVT) reaction and formulated for in vivo delivery. Mature mRNA requires a 5'-cap for gene expression and mRNA stability. There are two methods to add a cap in vitro: via a two-step multi-enzymatic reaction or co-transcriptionally. Co-transcriptional methods minimize reaction steps and enzymes needed to make mRNA when compared to enzymatic capping. CleanCap® AG co-transcriptional capping results in 5 mg/ml of IVT with 94% 5'-cap 1 structure. This is highly efficient compared to first-generation cap analogs, such as mCap and ARCA, that incorporate cap 0 structures at lower efficiency and reaction yield. This article describes co-transcriptional capping using TriLink Biotechnology's CleanCap® AG in IVT. © 2021 Wiley Periodicals LLC. Basic Protocol 1: IVT with CleanCap Basic Protocol 2: mRNA purification and analysis.


Subject(s)
RNA Cap Analogs/chemical synthesis , RNA, Messenger/chemical synthesis , Humans , In Vitro Techniques , Protein Biosynthesis , RNA Stability , RNA, Messenger/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL