1
|
Sinha MH, Mehtab T, Entesari M, Nguyen HH, Yun A, Hwang I. Production of active human iduronate-2-sulfatase (IDS) enzyme in Nicotiana benthamiana. Sci Rep 2024; 14:23066. [PMID: 39367006 PMCID: PMC11452390 DOI: 10.1038/s41598-024-73778-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/20/2024] [Indexed: 10/06/2024] Open
Abstract
Many strategies have been developed to produce high levels of biologically active recombinant proteins in plants for biopharmaceutical purposes. However, the production of an active form of human iduronate-2-sulfatase (hIDS) for the treatment of Hunter syndrome by enzyme replacement therapy (ERT) is challenging due to the requirement for cotranslational modification by a formylglycine-producing enzyme encoded by sulfatase modifying factor 1 (hSUMF1) at the Cys84 residue, which converts it to C(alpha)-formylglycine. In this study, we have shown that hIDS can be highly expressed in N. benthamiana by using different constructs. Among them, BiP-GB1-L-dCBD1-2L-8xHis-L-6xHis-3L-EK-hIDS-HDEL (GB1-CBD1-hIDS) showed a high expression level when transiently co-expressed with the turnip crinkle virus gene silencing suppressor P38 and GB1-fused human calreticulin (GB1-CRT1) as a folding enhancer. The hSUMF1 was co-expressed with hIDS for cotranslational modification. The full-length recombinant proteins were purified using Ni2+-NTA affinity resin followed by enterokinase treatment to obtain tag-free hIDS. The N-terminal fragment was removed using microcrystalline cellulose (MCC) beads. The purified active form of hIDS can successfully cleave the sulfate group from an artificial substrate, 4-nitrocatechol sulfate, at a similar level to commercial hIDS expressed in animal cells. These results suggest that plants could be a promising platform for the production of recombinant hIDS.
Collapse
Affiliation(s)
- Md Hasif Sinha
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Tahrin Mehtab
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Mehrnaz Entesari
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Hong Hanh Nguyen
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Areum Yun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea
| | - Inhwan Hwang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, South Korea.
| |
Collapse
|
2
|
Song SJ, Diao HP, Guo YF, Hwang I. Advances in Subcellular Accumulation Design for Recombinant Protein Production in Tobacco. BIODESIGN RESEARCH 2024; 6:0047. [PMID: 39206181 PMCID: PMC11350518 DOI: 10.34133/bdr.0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Plants and their use as bioreactors for the generation of recombinant proteins have become one of the hottest topics in the field of Plant Biotechnology and Plant Synthetic Biology. Plant bioreactors offer superior engineering potential compared to other types, particularly in the realm of subcellular accumulation strategies for increasing production yield and quality. This review explores established and emerging strategies for subcellular accumulation of recombinant proteins in tobacco bioreactors, highlighting recent advancements in the field. Additionally, the review provides reference to the crucial initial step of selecting an optimal subcellular localization for the target protein, a design that substantially impacts production outcomes.
Collapse
Affiliation(s)
- Shi-Jian Song
- Tobacco Research Institute,
Chinese Academy of Agricultural Sciences, Qingdao, China
- Beijing Life Science Academy (BLSA), Beijing, China
| | - Hai-Ping Diao
- Tobacco Research Institute,
Chinese Academy of Agricultural Sciences, Qingdao, China
| | - Yong-Feng Guo
- Tobacco Research Institute,
Chinese Academy of Agricultural Sciences, Qingdao, China
| | - Inhwan Hwang
- Department of Life Science,
Pohang University of Science and Technology, Pohang, Republic of Korea
- BioApplications Inc., Pohang, Republic of Korea
| |
Collapse
|
3
|
Torres-Herrero B, Armenia I, Ortiz C, de la Fuente JM, Betancor L, Grazú V. Opportunities for nanomaterials in enzyme therapy. J Control Release 2024; 372:619-647. [PMID: 38909702 DOI: 10.1016/j.jconrel.2024.06.035] [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: 03/10/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
In recent years, enzyme therapy strategies have rapidly evolved to catalyze essential biochemical reactions with therapeutic potential. These approaches hold particular promise in addressing rare genetic disorders, cancer treatment, neurodegenerative conditions, wound healing, inflammation management, and infectious disease control, among others. There are several primary reasons for the utilization of enzymes as therapeutics: their substrate specificity, their biological compatibility, and their ability to generate a high number of product molecules per enzyme unit. These features have encouraged their application in enzyme replacement therapy where the enzyme serves as the therapeutic agent to rectify abnormal metabolic and physiological processes, enzyme prodrug therapy where the enzyme initiates a clinical effect by activating prodrugs, and enzyme dynamic or starving therapy where the enzyme acts upon host substrate molecules. Currently, there are >20 commercialized products based on therapeutic enzymes, but approval rates are considerably lower than other biologicals. This has stimulated nanobiotechnology in the last years to develop nanoparticle-based solutions that integrate therapeutic enzymes. This approach aims to enhance stability, prevent rapid clearance, reduce immunogenicity, and even enable spatio-temporal activation of the therapeutic catalyst. This comprehensive review delves into emerging trends in the application of therapeutic enzymes, with a particular emphasis on the synergistic opportunities presented by incorporating enzymes into nanomaterials. Such integration holds the promise of enhancing existing therapies or even paving the way for innovative nanotherapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Torres-Herrero
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Cecilia Ortiz
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Jesús Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Lorena Betancor
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
4
|
Pimentel-Vera LN, Rodríguez-López A, Espejo-Mojica AJ, Ramírez AM, Cardona C, Reyes LH, Tomatsu S, Jaroentomeechai T, DeLisa MP, Sánchez OF, Alméciga-Díaz CJ. Novel human recombinant N-acetylgalactosamine-6-sulfate sulfatase produced in a glyco-engineered Escherichia coli strain. Heliyon 2024; 10:e32555. [PMID: 38952373 PMCID: PMC11215262 DOI: 10.1016/j.heliyon.2024.e32555] [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: 02/19/2024] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Abstract
Mucopolysaccharidosis IVA (MPS IVA) is a lysosomal storage disease caused by mutations in the gene encoding the lysosomal enzyme N-acetylgalactosamine-6-sulfate sulfatase (GALNS), resulting in the accumulation of keratan sulfate (KS) and chondroitin-6-sulfate (C6S). Previously, it was reported the production of an active human recombinant GALNS (rGALNS) in E. coli BL21(DE3). However, this recombinant enzyme was not taken up by HEK293 cells or MPS IVA skin fibroblasts. Here, we leveraged a glyco-engineered E. coli strain to produce a recombinant human GALNS bearing the eukaryotic trimannosyl core N-glycan, Man3GlcNAc2 (rGALNSoptGly). The N-glycosylated GALNS was produced at 100 mL and 1.65 L scales, purified and characterized with respect to pH stability, enzyme kinetic parameters, cell uptake, and KS clearance. The results showed that the addition of trimannosyl core N-glycans enhanced both protein stability and substrate affinity. rGALNSoptGly was capture through a mannose receptor-mediated process. This enzyme was delivered to the lysosome, where it reduced KS storage in human MPS IVA fibroblasts. This study demonstrates the potential of a glyco-engineered E. coli for producing a fully functional GALNS enzyme. It may offer an economic approach for the biosynthesis of a therapeutic glycoprotein that could prove useful for MPS IVA treatment. This strategy could be extended to other lysosomal enzymes that rely on the presence of mannose N-glycans for cell uptake.
Collapse
Affiliation(s)
- Luisa N. Pimentel-Vera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
| | - Alexander Rodríguez-López
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
- Dogma Biotech, Bogotá, D.C., 110111, Colombia
| | - Angela J. Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
- Dogma Biotech, Bogotá, D.C., 110111, Colombia
| | - Aura María Ramírez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
| | - Carolina Cardona
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
- Grupo de Investigaciones Biomédicas y de Genética Humana Aplicada GIBGA, Facultad de Ciencias de la Salud, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A, Bogotá, D.C., Colombia
| | - Luis H. Reyes
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá, D.C., Colombia
| | - Shunji Tomatsu
- Nemours Children's Health, Wilmington, DE, 19803, USA
- Faculty of Arts and Sciences, University of Delaware, Newark, DE, 19716, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, 19144, USA
| | - Thapakorn Jaroentomeechai
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Matthew P. DeLisa
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Cornell Institute of Biotechnology, Cornell University, Ithaca, NY, USA
| | - Oscar F. Sánchez
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Carlos J. Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá, D.C., 110231, Colombia
| |
Collapse
|
5
|
Itoh K, Tsukimoto J. Lysosomal sialidase NEU1, its intracellular properties, deficiency, and use as a therapeutic agent. Glycoconj J 2023; 40:611-619. [PMID: 38147151 DOI: 10.1007/s10719-023-10135-6] [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: 01/16/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 12/27/2023]
Abstract
Neuraminidase 1 (NEU1) is a lysosomal sialidase that cleaves terminal α-linked sialic acid residues from sialylglycans. NEU1 is biosynthesized in the rough endoplasmic reticulum (RER) lumen as an N-glycosylated protein to associate with its protective protein/cathepsin A (CTSA) and then form a lysosomal multienzyme complex (LMC) also containing β-galactosidase 1 (GLB1). Unlike other mammalian sialidases, including NEU2 to NEU4, NEU1 transport to lysosomes requires association of NEU1 with CTSA, binding of the CTSA carrying terminal mannose 6-phosphate (M6P)-type N-glycan with M6P receptor (M6PR), and intralysosomal NEU1 activation at acidic pH. In contrast, overexpression of the single NEU1 gene in mammalian cells causes intracellular NEU1 protein crystallization in the RER due to self-aggregation when intracellular CTSA is reduced to a relatively low level. Sialidosis (SiD) and galactosialidosis (GS) are autosomal recessive lysosomal storage diseases caused by the gene mutations of NEU1 and CTSA, respectively. These incurable diseases associate with the NEU1 deficiency, excessive accumulation of sialylglycans in neurovisceral organs, and systemic manifestations. We established a novel GS model mouse carrying homozygotic Ctsa IVS6 + 1 g/a mutation causing partial exon 6 skipping with simultaneous deficiency of Ctsa and Neu1. Symptoms developed in the GS mice like those in juvenile/adult GS patients, such as myoclonic seizures, suppressed behavior, gargoyle-like face, edema, proctoptosis due to Neu1 deficiency, and sialylglycan accumulation associated with neurovisceral inflammation. We developed a modified NEU1 (modNEU1), which does not form protein crystals but is transported to lysosomes by co-expressed CTSA. In vivo gene therapy for GS and SiD utilizing a single adeno-associated virus (AAV) carrying modNEU1 and CTSA genes under dual promoter control will be created.
Collapse
Affiliation(s)
- Kohji Itoh
- Department of Medicinal Biotechnology, Institute for Medicinal Biotechnology, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan.
| | - Jun Tsukimoto
- Department of Medicinal Biotechnology, Institute for Medicinal Biotechnology, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
6
|
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: 10] [Impact Index Per Article: 10.0] [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
|
7
|
Buyel JF. Product safety aspects of plant molecular farming. Front Bioeng Biotechnol 2023; 11:1238917. [PMID: 37614627 PMCID: PMC10442644 DOI: 10.3389/fbioe.2023.1238917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
Plant molecular farming (PMF) has been promoted since the 1990s as a rapid, cost-effective and (most of all) safe alternative to the cultivation of bacteria or animal cells for the production of biopharmaceutical proteins. Numerous plant species have been investigated for the production of a broad range of protein-based drug candidates. The inherent safety of these products is frequently highlighted as an advantage of PMF because plant viruses do not replicate in humans and vice versa. However, a more nuanced analysis of this principle is required when considering other pathogens because toxic compounds pose a risk even in the absence of replication. Similarly, it is necessary to assess the risks associated with the host system (e.g., the presence of toxic secondary metabolites) and the production approach (e.g., transient expression based on bacterial infiltration substantially increases the endotoxin load). This review considers the most relevant host systems in terms of their toxicity profile, including the presence of secondary metabolites, and the risks arising from the persistence of these substances after downstream processing and product purification. Similarly, we discuss a range of plant pathogens and disease vectors that can influence product safety, for example, due to the release of toxins. The ability of downstream unit operations to remove contaminants and process-related toxic impurities such as endotoxins is also addressed. This overview of plant-based production, focusing on product safety aspects, provides recommendations that will allow stakeholders to choose the most appropriate strategies for process development.
Collapse
Affiliation(s)
- J. F. Buyel
- Department of Biotechnology (DBT), Institute of Bioprocess Science and Engineering (IBSE), University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| |
Collapse
|
8
|
Isolation of Salvia miltiorrhiza Kaurene Synthase-like ( KSL) Gene Promoter and Its Regulation by Ethephon and Yeast Extract. Genes (Basel) 2022; 14:genes14010054. [PMID: 36672795 PMCID: PMC9859234 DOI: 10.3390/genes14010054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
The presented study describes the regulation of the promoter region of the Salvia miltiorrhiza kaurene synthase-like gene (SmKSL) by ethylene and yeast extract. The isolated fragment is 897 bp and is composed of a promoter (763 bp), 5'UTR (109 bp), and a short CDS (25 bp). The initial in silico analysis revealed the presence of numerous putative cis-active sites for trans-factors responding to different stress conditions. However, this study examines the influence of ethylene and yeast extract on SmKSL gene expression and tanshinone biosynthesis regulation. The results of 72h RT-PCR indicate an antagonistic interaction between ethylene, provided as ethephon (0.05, 0.10, 0.25, and 0.50 mM), and yeast extract (0.5%) on SmKSL gene expression in callus cultures of S. miltiorrhiza. A similar antagonistic effect was observed on total tanshinone concentration for up to 60 days. Ethylene provided as ethephon (0.05, 0.10, 0.25, and 0.50 mM) is a weak inducer of total tanshinone biosynthesis, increasing them only up to the maximum value of 0.67 ± 0.04 mg g-1 DW (60-day induction with 0.50 mM ethephon). Among the tanshinones elicited by ethephon, cryptotanshinone (52.21%) dominates, followed by dihydrotanshinone (45.00%) and tanshinone IIA (3.79%). In contrast, the 0.5% yeast extract strongly increases the total tanshinone concentration up to a maximum value of 13.30 ± 1.09 mg g-1 DW, observed after 50 days of induction. Yeast extract and ethylene appear to activate different fragments of the tanshinone biosynthesis route; hence the primary tanshinones induced by yeast extract were cryptotanshinone (81.42%), followed by dihydrotanshinone (17.06%) and tanshinone IIA (1.52%).
Collapse
|
9
|
Kulshreshtha A, Sharma S, Padilla CS, Mandadi KK. Plant-based expression platforms to produce high-value metabolites and proteins. FRONTIERS IN PLANT SCIENCE 2022; 13:1043478. [PMID: 36426139 PMCID: PMC9679013 DOI: 10.3389/fpls.2022.1043478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Plant-based heterologous expression systems can be leveraged to produce high-value therapeutics, industrially important proteins, metabolites, and bioproducts. The production can be scaled up, free from pathogen contamination, and offer post-translational modifications to synthesize complex proteins. With advancements in molecular techniques, transgenics, CRISPR/Cas9 system, plant cell, tissue, and organ culture, significant progress has been made to increase the expression of recombinant proteins and important metabolites in plants. Methods are also available to stabilize RNA transcripts, optimize protein translation, engineer proteins for their stability, and target proteins to subcellular locations best suited for their accumulation. This mini-review focuses on recent advancements to enhance the production of high-value metabolites and proteins necessary for therapeutic applications using plants as bio-factories.
Collapse
Affiliation(s)
- Aditya Kulshreshtha
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
| | - Shweta Sharma
- Department of Veterinary Pathology, Dr. GCN College of Veterinary & Animal Sciences, CSK Himachal Pradesh Agricultural University, Palampur, India
| | - Carmen S. Padilla
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
| | - Kranthi K. Mandadi
- Texas A&M AgriLife Research and Extension Center, Weslaco, TX, United States
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, United States
- Institute for Advancing Health Through Agriculture, Texas A&M AgriLife, College Station, TX, United States
| |
Collapse
|
10
|
Chen J, Huang Y, Shu Y, Hu X, Wu D, Jiang H, Wang K, Liu W, Fu W. Recent Progress on Systems and Synthetic Biology of Diatoms for Improving Algal Productivity. Front Bioeng Biotechnol 2022; 10:908804. [PMID: 35646842 PMCID: PMC9136054 DOI: 10.3389/fbioe.2022.908804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Microalgae have drawn much attention for their potential applications as a sustainable source for developing bioactive compounds, functional foods, feeds, and biofuels. Diatoms, as one major group of microalgae with high yields and strong adaptability to the environment, have shown advantages in developing photosynthetic cell factories to produce value-added compounds, including heterologous bioactive products. However, the commercialization of diatoms has encountered several obstacles that limit the potential mass production, such as the limitation of algal productivity and low photosynthetic efficiency. In recent years, systems and synthetic biology have dramatically improved the efficiency of diatom cell factories. In this review, we discussed first the genome sequencing and genome-scale metabolic models (GEMs) of diatoms. Then, approaches to optimizing photosynthetic efficiency are introduced with a focus on the enhancement of biomass productivity in diatoms. We also reviewed genome engineering technologies, including CRISPR (clustered regularly interspaced short palindromic repeats) gene-editing to produce bioactive compounds in diatoms. Finally, we summarized the recent progress on the diatom cell factory for producing heterologous compounds through genome engineering to introduce foreign genes into host diatoms. This review also pinpointed the bottlenecks in algal engineering development and provided critical insights into the future direction of algal production.
Collapse
Affiliation(s)
- Jiwei Chen
- Department of Marine Science, Ocean College, Zhejiang University, Hangzhou, China
| | - Yifan Huang
- Department of Marine Science, Ocean College, Zhejiang University, Hangzhou, China
| | - Yuexuan Shu
- Department of Marine Science, Ocean College, Zhejiang University, Hangzhou, China
| | - Xiaoyue Hu
- Center for Data Science, Zhejiang University, Hangzhou, China
- School of Mathematical Sciences, Zhejiang University, Hangzhou, China
| | - Di Wu
- Department of Marine Science, Ocean College, Zhejiang University, Hangzhou, China
| | - Hangjin Jiang
- Center for Data Science, Zhejiang University, Hangzhou, China
| | - Kui Wang
- Department of Marine Science, Ocean College, Zhejiang University, Hangzhou, China
| | - Weihua Liu
- School of Mathematical Sciences, Zhejiang University, Hangzhou, China
| | - Weiqi Fu
- Department of Marine Science, Ocean College, Zhejiang University, Hangzhou, China
- Center for Systems Biology and Faculty of Industrial Engineering, Mechanical Engineering and Computer Science, School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
- *Correspondence: Weiqi Fu,
| |
Collapse
|
11
|
van der Kaaij A, van Noort K, Nibbering P, Wilbers RHP, Schots A. Glyco-Engineering Plants to Produce Helminth Glycoproteins as Prospective Biopharmaceuticals: Recent Advances, Challenges and Future Prospects. FRONTIERS IN PLANT SCIENCE 2022; 13:882835. [PMID: 35574113 PMCID: PMC9100689 DOI: 10.3389/fpls.2022.882835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/28/2022] [Indexed: 06/15/2023]
Abstract
Glycoproteins are the dominant category among approved biopharmaceuticals, indicating their importance as therapeutic proteins. Glycoproteins are decorated with carbohydrate structures (or glycans) in a process called glycosylation. Glycosylation is a post-translational modification that is present in all kingdoms of life, albeit with differences in core modifications, terminal glycan structures, and incorporation of different sugar residues. Glycans play pivotal roles in many biological processes and can impact the efficacy of therapeutic glycoproteins. The majority of biopharmaceuticals are based on human glycoproteins, but non-human glycoproteins, originating from for instance parasitic worms (helminths), form an untapped pool of potential therapeutics for immune-related diseases and vaccine candidates. The production of sufficient quantities of correctly glycosylated putative therapeutic helminth proteins is often challenging and requires extensive engineering of the glycosylation pathway. Therefore, a flexible glycoprotein production system is required that allows straightforward introduction of heterologous glycosylation machinery composed of glycosyltransferases and glycosidases to obtain desired glycan structures. The glycome of plants creates an ideal starting point for N- and O-glyco-engineering of helminth glycans. Plants are also tolerant toward the introduction of heterologous glycosylation enzymes as well as the obtained glycans. Thus, a potent production platform emerges that enables the production of recombinant helminth proteins with unusual glycans. In this review, we discuss recent advances in plant glyco-engineering of potentially therapeutic helminth glycoproteins, challenges and their future prospects.
Collapse
|
12
|
Long Y, Wei X, Wu S, Wu N, Li QX, Tan B, Wan X. Plant Molecular Farming, a Tool for Functional Food Production. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:2108-2116. [PMID: 35139640 DOI: 10.1021/acs.jafc.1c07185] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The demand of functional food is increasing for improving human health. Plant molecular farming (PMF) employs plants as bioreactors for the production of pharmaceuticals. Now PMF has been used to produce antibodies, vaccines, and medicinal proteins, but it has not been well-studied for production of nutraceuticals and functional food. In this perspective, we extend the concept of PMF, present an updated overview of PMF for functional food development, including the progress, problem, and strategy, and then speculate how to use the PMF strategy to produce functional foods, especially with four major staple food crops (rice, wheat, maize, and soybean). Finally, we discuss the opportunities and challenges of PMF on functional food production in the future.
Collapse
Affiliation(s)
- Yan Long
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing, Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| | - Xun Wei
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing, Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| | - Suowei Wu
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing, Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| | - Nana Wu
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, People's Republic of China
| | - Qing X Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawaii 96822, United States
| | - Bin Tan
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, People's Republic of China
- School of Food Engineering, Harbin University of Commerce, Harbin, Heilongjiang 150076, People's Republic of China
| | - Xiangyuan Wan
- Zhongzhi International Institute of Agricultural Biosciences, Shunde Graduate School, Research Center of Biology and Agriculture, University of Science and Technology Beijing, Beijing 100024, People's Republic of China
- Beijing Beike Institute of Precision Medicine and Health Technology, Beijing 100192, People's Republic of China
- Beijing Engineering Laboratory of Main Crop Bio-Tech Breeding, Beijing International Science and Technology Cooperation Base of Bio-Tech Breeding, Beijing Solidwill Sci-Tech Company, Limited, Beijing 100192, People's Republic of China
| |
Collapse
|
13
|
Bohlender LL, Parsons J, Hoernstein SNW, Bangert N, Rodríguez-Jahnke F, Reski R, Decker EL. Unexpected Arabinosylation after Humanization of Plant Protein N-Glycosylation. Front Bioeng Biotechnol 2022; 10:838365. [PMID: 35252146 PMCID: PMC8894861 DOI: 10.3389/fbioe.2022.838365] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/28/2022] [Indexed: 02/03/2023] Open
Abstract
As biopharmaceuticals, recombinant proteins have become indispensable tools in medicine. An increasing demand, not only in quantity but also in diversity, drives the constant development and improvement of production platforms. The N-glycosylation pattern on biopharmaceuticals plays an important role in activity, serum half-life and immunogenicity. Therefore, production platforms with tailored protein N-glycosylation are of great interest. Plant-based systems have already demonstrated their potential to produce pharmaceutically relevant recombinant proteins, although their N-glycan patterns differ from those in humans. Plants have shown great plasticity towards the manipulation of their glycosylation machinery, and some have already been glyco-engineered in order to avoid the attachment of plant-typical, putatively immunogenic sugar residues. This resulted in complex-type N-glycans with a core structure identical to the human one. Compared to humans, plants lack the ability to elongate these N-glycans with β1,4-linked galactoses and terminal sialic acids. However, these modifications, which require the activity of several mammalian enzymes, have already been achieved for Nicotiana benthamiana and the moss Physcomitrella. Here, we present the first step towards sialylation of recombinant glycoproteins in Physcomitrella, human β1,4-linked terminal N-glycan galactosylation, which was achieved by the introduction of a chimeric β1,4-galactosyltransferase (FTGT). This chimeric enzyme consists of the moss α1,4-fucosyltransferase transmembrane domain, fused to the catalytic domain of the human β1,4-galactosyltransferase. Stable FTGT expression led to the desired β1,4-galactosylation. However, additional pentoses of unknown identity were also observed. The nature of these pentoses was subsequently determined by Western blot and enzymatic digestion followed by mass spectrometric analysis and resulted in their identification as α-linked arabinoses. Since a pentosylation of β1,4-galactosylated N-glycans was reported earlier, e.g., on recombinant human erythropoietin produced in glyco-engineered Nicotiana tabacum, this phenomenon is of a more general importance for plant-based production platforms. Arabinoses, which are absent in humans, may prevent the full humanization of plant-derived products. Therefore, the identification of these pentoses as arabinoses is important as it creates the basis for their abolishment to ensure the production of safe biopharmaceuticals in plant-based systems.
Collapse
Affiliation(s)
- Lennard L. Bohlender
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Juliana Parsons
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | | | - Nina Bangert
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Fernando Rodríguez-Jahnke
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Ralf Reski
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Eva L. Decker
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- *Correspondence: Eva L. Decker,
| |
Collapse
|
14
|
Recent advances in molecular farming using monocot plants. Biotechnol Adv 2022; 58:107913. [DOI: 10.1016/j.biotechadv.2022.107913] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/13/2022] [Accepted: 01/15/2022] [Indexed: 12/22/2022]
|
15
|
Dumitrascu DL. Gaucher disease: an update. Med Pharm Rep 2021; 94:S54-S56. [PMID: 34527912 DOI: 10.15386/mpr-2231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Gaucher disease is a lysosomal storage disease affecting the bone marrow, spleen, liver, and nervous system. In Romania we follow up over 70 adult patients with Gaucher disease, who benefit from fully covered therapy. There is a need to screen for Gaucher disease, to diagnose early the condition and to use the best available therapy. This is a review of recent studies on Gaucher disease and is dedicated to trainees in medicine.
Collapse
Affiliation(s)
- Dan L Dumitrascu
- 2Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
16
|
Ward BJ, Gobeil P, Séguin A, Atkins J, Boulay I, Charbonneau PY, Couture M, D'Aoust MA, Dhaliwall J, Finkle C, Hager K, Mahmood A, Makarkov A, Cheng MP, Pillet S, Schimke P, St-Martin S, Trépanier S, Landry N. Phase 1 randomized trial of a plant-derived virus-like particle vaccine for COVID-19. Nat Med 2021; 27:1071-1078. [PMID: 34007070 PMCID: PMC8205852 DOI: 10.1038/s41591-021-01370-1] [Citation(s) in RCA: 190] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Several severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are being deployed, but the global need greatly exceeds the supply, and different formulations might be required for specific populations. Here we report Day 42 interim safety and immunogenicity data from an observer-blinded, dose escalation, randomized controlled study of a virus-like particle vaccine candidate produced in plants that displays the SARS-CoV-2 spike glycoprotein (CoVLP: NCT04450004). The co-primary outcomes were the short-term tolerability/safety and immunogenicity of CoVLP formulations assessed by neutralizing antibody (NAb) and cellular responses. Secondary outcomes in this ongoing study include safety and immunogenicity assessments up to 12 months after vaccination. Adults (18–55 years, n = 180) were randomized at two sites in Quebec, Canada, to receive two intramuscular doses of CoVLP (3.75 μg, 7.5 μg, and 15 μg) 21 d apart, alone or adjuvanted with AS03 or CpG1018. All formulations were well tolerated, and adverse events after vaccination were generally mild to moderate, transient and highest in the adjuvanted groups. There was no CoVLP dose effect on serum NAbs, but titers increased significantly with both adjuvants. After the second dose, NAbs in the CoVLP + AS03 groups were more than tenfold higher than titers in Coronavirus 2019 convalescent sera. Both spike protein-specific interferon-γ and interleukin-4 cellular responses were also induced. This pre-specified interim analysis supports further evaluation of the CoVLP vaccine candidate. Safety and immunogenicity results in humans of a two-dose SARS-CoV-2 vaccine made from plants support further assessment of potential efficacy.
Collapse
Affiliation(s)
- Brian J Ward
- Medicago Inc., Quebec City, Quebec, Canada. .,Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | | - Matthew P Cheng
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
17
|
Massaro G, Geard AF, Liu W, Coombe-Tennant O, Waddington SN, Baruteau J, Gissen P, Rahim AA. Gene Therapy for Lysosomal Storage Disorders: Ongoing Studies and Clinical Development. Biomolecules 2021; 11:611. [PMID: 33924076 PMCID: PMC8074255 DOI: 10.3390/biom11040611] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Rare monogenic disorders such as lysosomal diseases have been at the forefront in the development of novel treatments where therapeutic options are either limited or unavailable. The increasing number of successful pre-clinical and clinical studies in the last decade demonstrates that gene therapy represents a feasible option to address the unmet medical need of these patients. This article provides a comprehensive overview of the current state of the field, reviewing the most used viral gene delivery vectors in the context of lysosomal storage disorders, a selection of relevant pre-clinical studies and ongoing clinical trials within recent years.
Collapse
Affiliation(s)
- Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Amy F. Geard
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Wenfei Liu
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Oliver Coombe-Tennant
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| | - Simon N. Waddington
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
- Gene Transfer Technology Group, EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK
| | - Julien Baruteau
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK;
- Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, National Institute of Health Research, University College London, London WC1N 1EH, UK;
| | - Paul Gissen
- Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, National Institute of Health Research, University College London, London WC1N 1EH, UK;
| | - Ahad A. Rahim
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK; (A.F.G.); (W.L.); (O.C.-T.); (A.A.R.)
| |
Collapse
|
18
|
Göritzer K, Strasser R. Glycosylation of Plant-Produced Immunoglobulins. EXPERIENTIA SUPPLEMENTUM (2012) 2021; 112:519-543. [PMID: 34687021 DOI: 10.1007/978-3-030-76912-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Many economically important protein-based therapeutics like monoclonal antibodies are glycosylated. Due to the recognized importance of this type of posttranslational modification, glycoengineering of expression systems to obtain highly active and homogenous therapeutics is an emerging field. Although most of the monoclonal antibodies on the market are still produced in mammalian expression platforms, plants are emerging as an alternative cost-effective and scalable production platform that allows precise engineering of glycosylation to produce targeted human glycoforms at large homogeneity. Apart from producing more effective antibodies, pure glycoforms are required in efforts to link biological functions to specific glycan structures. Much is already known about the role of IgG1 glycosylation and this antibody class is the dominant recombinant format that has been expressed in plants. By contrast, little attention has been paid to the glycoengineering of recombinant IgG subtypes and the other four classes of human immunoglobulins (IgA, IgD, IgE, and IgM). Except for IgD, all these antibody classes have been expressed in plants and the glycosylation has been analyzed in a site-specific manner. Here, we summarize the current data on glycosylation of plant-produced monoclonal antibodies and discuss the findings in the light of known functions for these glycans.
Collapse
Affiliation(s)
| | - Richard Strasser
- University of Natural Resources and Life Sciences Vienna, Vienna, Austria.
| |
Collapse
|
19
|
Uthailak N, Kajiura H, Misaki R, Fujiyama K. Transient Production of Human β-Glucocerebrosidase With Mannosidic-Type N-Glycan Structure in Glycoengineered Nicotiana benthamiana Plants. FRONTIERS IN PLANT SCIENCE 2021; 12:683762. [PMID: 34163514 PMCID: PMC8215604 DOI: 10.3389/fpls.2021.683762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/07/2021] [Indexed: 05/02/2023]
Abstract
Gaucher disease is an inherited lysosomal storage disorder caused by a deficiency of functional enzyme β-glucocerebrosidase (GCase). Recombinant GCase has been used in enzyme replacement therapy to treat Gaucher disease. Importantly, the terminal mannose N-glycan structure is essential for the uptake of recombinant GCase into macrophages via the mannose receptor. In this research, recombinant GCase was produced using Agrobacterium-mediated transient expression in both wild-type (WT) and N-acetylglucosaminyltransferase I (GnTI) downregulated Nicotiana benthamiana (ΔgntI) plants, the latter of which accumulates mannosidic-type N-glycan structures. The successfully produced functional GCase exhibited GCase enzyme activity. The enzyme activity was the same as that of the conventional mammalian-derived GCase. Notably, N-glycan analysis revealed that a mannosidic-type N-glycan structure lacking plant-specific N-glycans (β1,2-xylose and α1,3-fucose residues) was predominant in all glycosylation sites of purified GCase produced from ΔgntI plants. Our research provides a promising alternative plant line as a host for the production of recombinant GCase with a mannosidic-type N-glycan structure. This glycoengineered plant might be applicable to the production of other pharmaceutical proteins, especially mannose receptor targeted protein, for therapeutic uses.
Collapse
Affiliation(s)
| | - Hiroyuki Kajiura
- International Center for Biotechnology, Osaka University, Osaka, Japan
- Industrial Biotechnology Initiative Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Ryo Misaki
- International Center for Biotechnology, Osaka University, Osaka, Japan
- Industrial Biotechnology Initiative Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Kazuhito Fujiyama
- International Center for Biotechnology, Osaka University, Osaka, Japan
- Industrial Biotechnology Initiative Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Cooperative Research Station in Southeast Asia, International Center for Biotechnology, Osaka University, Mahidol University, Bangkok, Thailand
- *Correspondence: Kazuhito Fujiyama
| |
Collapse
|
20
|
Mahmood N, Nasir SB, Hefferon K. Plant-Based Drugs and Vaccines for COVID-19. Vaccines (Basel) 2020; 9:15. [PMID: 33396667 PMCID: PMC7823519 DOI: 10.3390/vaccines9010015] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 01/08/2023] Open
Abstract
The coronavirus SARS-CoV-2 has turned our own health and the world economy upside down. While several vaccine candidates are currently under development, antivirals with the potential to limit virus transmission or block infection are also being explored. Plant production platforms are being used to generate vaccines and antiviral proteins inexpensively and at mass scale. The following review discusses the biology and origins of the current coronavirus pandemic, and describes some of the conventional, synthetic, and plant-based approaches to address the challenge that it presents to our way of life.
Collapse
Affiliation(s)
- Nasir Mahmood
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 2E8, Canada;
- Department of Biochemistry, University of Health Sciences, Lahore 54600, Pakistan
- Forest Ridge Health Care Inc., Toronto, ON M5J 2V1, Canada
| | - Sarah Bushra Nasir
- Department of Life Sciences, Abdus Salam School of Sciences, Nusrat Jahan College, Chenab Nagar 35460, Pakistan;
| | | |
Collapse
|
21
|
Huebbers JW, Buyel JF. On the verge of the market - Plant factories for the automated and standardized production of biopharmaceuticals. Biotechnol Adv 2020; 46:107681. [PMID: 33326816 DOI: 10.1016/j.biotechadv.2020.107681] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/19/2020] [Accepted: 12/08/2020] [Indexed: 12/28/2022]
Abstract
The market for biopharmaceuticals is dominated by recombinant proteins and is driven mainly by the development of vaccines and antibodies. Manufacturing predominantly relies on fermentation-based production platforms, which have limited scalability and suffer from high upstream process costs. As an alternative, the production of recombinant proteins in whole plants (plant molecular farming) provides a scalable and cost efficient upstream process because each plant functions as a self-contained bioreactor, avoiding costs associated with single-use devices and cleaning-in-place. Despite many proof-of-concept studies and the approval of a few products as medical devices, the only approved pharmaceutical proteins manufactured in whole plants have been authorized under emergency protocols. The absence of approvals under standard clinical development pathways in part reflects the lack of standardized process equipment and unit operations, leading to industry inertia based on familiarity with fermenter systems. Here we discuss the upstream production steps of plant molecular farming by transient expression in intact plants, including seeding, plant cultivation, infiltration with Agrobacterium tumefaciens, post-infiltration incubation, and harvesting. We focus on cultivation techniques because they strongly affect the subsequent steps and overall process design. We compare the benefits and drawbacks of open field, greenhouse and vertical farm strategies in terms of upfront investment costs, batch reproducibility, and decoupling from environmental impacts. We consider process automation, monitoring and adaptive process design in the context of Industry 4.0, which can boost process efficiency and batch-to-batch uniformity to improve regulatory compliance. Finally, we discuss the costs-benefit aspects of the different cultivation systems.
Collapse
Affiliation(s)
- J W Huebbers
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074 Aachen, Germany.
| | - J F Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074 Aachen, Germany; Institute for Molecular Biotechnology, Worringerweg 1, RWTH Aachen University, 52074 Aachen, Germany.
| |
Collapse
|
22
|
Srikanth MP, Feldman RA. Elevated Dkk1 Mediates Downregulation of the Canonical Wnt Pathway and Lysosomal Loss in an iPSC Model of Neuronopathic Gaucher Disease. Biomolecules 2020; 10:E1630. [PMID: 33287247 PMCID: PMC7761665 DOI: 10.3390/biom10121630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Gaucher Disease (GD), which is the most common lysosomal storage disorder, is caused by bi-allelic mutations in GBA1-a gene that encodes the lysosomal hydrolase β-glucocerebrosidase (GCase). The neuronopathic forms of GD (nGD) are characterized by severe neurological abnormalities that arise during gestation or early in infancy. Using GD-induced pluripotent stem cell (iPSC)-derived neuronal progenitor cells (NPCs), we have previously reported that neuronal cells have neurodevelopmental defects associated with the downregulation of canonical Wnt signaling. In this study, we report that GD NPCs display elevated levels of Dkk1, which is a secreted Wnt antagonist that prevents receptor activation. Dkk1 upregulation in mutant NPCs resulted in an increased degradation of β-catenin, and there was a concomitant reduction in lysosomal numbers. Consistent with these results, incubation of the mutant NPCs with recombinant Wnt3a (rWnt3a) was able to outcompete the excess Dkk1, increasing β-catenin levels and rescuing lysosomal numbers. Furthermore, the incubation of WT NPCs with recombinant Dkk1 (rDkk1) phenocopied the mutant phenotype, recapitulating the decrease in β-catenin levels and lysosomal depletion seen in nGD NPCs. This study provides evidence that downregulation of the Wnt/β-catenin pathway in nGD neuronal cells involves the upregulation of Dkk1. As Dkk1 is an extracellular Wnt antagonist, our results suggest that the deleterious effects of Wnt/β-catenin downregulation in nGD may be ameliorated by the prevention of Dkk1 binding to the Wnt co-receptor LRP6, pointing to Dkk1 as a potential therapeutic target for GBA1-associated neurodegeneration.
Collapse
Affiliation(s)
| | - Ricardo A. Feldman
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
| |
Collapse
|
23
|
O’Flaherty R, Bergin A, Flampouri E, Mota LM, Obaidi I, Quigley A, Xie Y, Butler M. Mammalian cell culture for production of recombinant proteins: A review of the critical steps in their biomanufacturing. Biotechnol Adv 2020; 43:107552. [DOI: 10.1016/j.biotechadv.2020.107552] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 12/28/2022]
|
24
|
Corbin JM, McNulty MJ, Macharoen K, McDonald KA, Nandi S. Technoeconomic analysis of semicontinuous bioreactor production of biopharmaceuticals in transgenic rice cell suspension cultures. Biotechnol Bioeng 2020; 117:3053-3065. [PMID: 32592492 DOI: 10.1002/bit.27475] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022]
Abstract
Biopharmaceutical protein production using transgenic plant cell bioreactor processes offers advantages over microbial and mammalian cell culture platforms in its ability to produce complex biologics with simple chemically defined media and reduced biosafety concerns. A disadvantage of plant cells from a traditional batch bioprocessing perspective is their slow growth rate which has motivated us to develop semicontinuous and/or perfusion processes. Although the economic benefits of plant cell culture bioprocesses are often mentioned in the literature, to our knowledge no rigorous technoeconomic models or analyses have been published. Here we present technoeconomic models in SuperPro Designer® for the large-scale production of recombinant butyrylcholinesterase (BChE), a prophylactic/therapeutic bioscavenger against organophosphate nerve agent poisoning, in inducible transgenic rice cell suspension cultures. The base facility designed to produce 25 kg BChE per year utilizing two-stage semicontinuous bioreactor operation manufactures a single 400 mg dose of BChE for $263. Semicontinuous operation scenarios result in 4-11% reduction over traditional two-stage batch operation scenarios. In addition to providing a simulation tool that will be useful to the plant-made pharmaceutical community, the model also provides a computational framework that can be used for other semicontinuous or batch bioreactor-based processes.
Collapse
Affiliation(s)
- Jasmine M Corbin
- Department of Chemical Engineering, University of California, Davis, California
| | - Matthew J McNulty
- Department of Chemical Engineering, University of California, Davis, California
| | | | - Karen A McDonald
- Department of Chemical Engineering, University of California, Davis, California.,Department of Molecular and Cellular Biology, Global HealthShare Initiative, University of California, Davis, California
| | - Somen Nandi
- Department of Chemical Engineering, University of California, Davis, California.,Department of Molecular and Cellular Biology, Global HealthShare Initiative, University of California, Davis, California
| |
Collapse
|
25
|
Datta S, Rajnish KN, George Priya Doss C, Melvin Samuel S, Selvarajan E, Zayed H. Enzyme therapy: a forerunner in catalyzing a healthy society? Expert Opin Biol Ther 2020; 20:1151-1174. [PMID: 32597245 DOI: 10.1080/14712598.2020.1787980] [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] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The use of enzymes in various industries has been prevalent for centuries. However, their potency as therapeutics remained latent until the late 1950 s, when scientists finally realized the gold mine they were sitting on. Enzyme therapy has seen rapid development over the past few decades and has been widely used for the therapy of myriad diseases, including lysosomal storage disorders, cancer, Alzheimer's disease, irritable bowel syndrome, exocrine pancreatic insufficiency, and hyperuricemia. Enzymes are also used for wound healing, the treatment of microbial infections, and gene therapy. AREAS COVERED This is a comprehensive review of the therapeutic use of enzymes that can act as a guidepost for researchers and academicians and presents a general overview of the developments in enzyme therapy over the years, along with updates on recent advancements in enzyme therapy research. EXPERT OPINION Although enzyme therapy is immensely beneficial and induces little auxiliary damage, it has several drawbacks, ranging from high cost, low stability, low production, and hyperimmune responses to the failure to cure a variety of the problems associated with a disease. Further fine-tuning and additional clinical efficacy studies are required to establish enzyme therapy as a forerunner to catalyzing a healthy society.
Collapse
Affiliation(s)
- Saptashwa Datta
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology , Kattankulathur, TN, India
| | - K Narayanan Rajnish
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology , Kattankulathur, TN, India
| | - C George Priya Doss
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology , Vellore, TN, India
| | - S Melvin Samuel
- Materials Science and Engineering, University of Wisconsin-Milwaukee , Milwaukee, WI, United States
| | - E Selvarajan
- Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology , Kattankulathur, TN, India
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health and Sciences, QU Health, Qatar University , Doha, Qatar
| |
Collapse
|
26
|
Towards a new avenue for producing therapeutic proteins: Microalgae as a tempting green biofactory. Biotechnol Adv 2020; 40:107499. [DOI: 10.1016/j.biotechadv.2019.107499] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/02/2019] [Accepted: 12/17/2019] [Indexed: 02/08/2023]
|
27
|
Shanmugaraj B, Malla A, Phoolcharoen W. Emergence of Novel Coronavirus 2019-nCoV: Need for Rapid Vaccine and Biologics Development. Pathogens 2020; 9:E148. [PMID: 32098302 PMCID: PMC7168632 DOI: 10.3390/pathogens9020148] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/24/2022] Open
Abstract
Novel Coronavirus (2019-nCoV) is an emerging pathogen that was first identified in Wuhan, China in late December 2019. This virus is responsible for the ongoing outbreak that causes severe respiratory illness and pneumonia-like infection in humans. Due to the increasing number of cases in China and outside China, the WHO declared coronavirus as a global health emergency. Nearly 35,000 cases were reported and at least 24 other countries or territories have reported coronavirus cases as early on as February. Inter-human transmission was reported in a few countries, including the United States. Neither an effective anti-viral nor a vaccine is currently available to treat this infection. As the virus is a newly emerging pathogen, many questions remain unanswered regarding the virus's reservoirs, pathogenesis, transmissibility, and much more is unknown. The collaborative efforts of researchers are needed to fill the knowledge gaps about this new virus, to develop the proper diagnostic tools, and effective treatment to combat this infection. Recent advancements in plant biotechnology proved that plants have the ability to produce vaccines or biopharmaceuticals rapidly in a short time. In this review, the outbreak of 2019-nCoV in China, the need for rapid vaccine development, and the potential of a plant system for biopharmaceutical development are discussed.
Collapse
Affiliation(s)
- Balamurugan Shanmugaraj
- Research unit for Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; (B.S.); (A.M.)
- Department of Pharmacognosy and Pharmaceutical Botany, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ashwini Malla
- Research unit for Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; (B.S.); (A.M.)
- Department of Pharmacognosy and Pharmaceutical Botany, Chulalongkorn University, Bangkok 10330, Thailand
| | - Waranyoo Phoolcharoen
- Research unit for Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand; (B.S.); (A.M.)
- Department of Pharmacognosy and Pharmaceutical Botany, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
28
|
Kim J, Do J, Choi HY, Kim SD, Park H, You S, Kim W, Jang Y, Kim D, Lee J, Ha J, Ji M, Kim DI, Kim HH. Profiles of plant core-fucosylated N-glycans of acid alpha-glucosidases produced in transgenic rice cell suspension cultures treated with eight different conditions. Enzyme Microb Technol 2020; 134:109482. [PMID: 32044029 DOI: 10.1016/j.enzmictec.2019.109482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/15/2019] [Accepted: 11/28/2019] [Indexed: 11/28/2022]
Abstract
Recombinant human acid alpha-glucosidase (rhGAA) from Chinese hamster ovary cells is the only approved treatment for patients with Pompe disease. In this study, rhGAAs were produced in transgenic rice cell suspension cultures under eight different conditions; untreated, 5 μM of 2-fluoro-l-fucose (2-FF), 50 μM of 2-FF, 100 μM of 2-FF, 100 μM of 2-FF + 0.5% Pluronic F-68 (PF-68), 100 μM of 2-FF + 0.05% Tween 20 (Tw 20), 0.5% PF-68, and 0.05% Tw 20. The N-glycans of eight rhGAAs were analyzed using ultra-performance liquid chromatography (UPLC) and tandem mass spectrometry. The relative quantity (%) of each glycan was obtained from the corresponding UPLC peak area per the sum (100%) of individual UPLC peak area. Fifteen N-glycans, comprising seven core-fucosylated glycans (71.5%, sum of each relative quantities) that have immunogenicity-inducing potential, three de-core-fucosylated glycans (15.4%), and five non-core-fucosylated glycans (13.1%), were characterized with high mass accuracy and glycan-generated fragment ions. The increases or decreases of relative quantities of each glycan from seven rhGAAs were compared with those of untreated control. The percentages of the sum of the relative quantities of core-fucosylated glycans divided by the sums of those of de-core- (core-fucose removed) and non-core-fucosylated glycans were calculated, and the lowest percentage was obtained in 100 μM of 2-FF combined with 0.5% PF-68. These results indicate that the relative quantity of each glycan of rhGAA produced in rice cell suspension cultures is significantly affected by their culture condition. This study performed the comparison of the N-glycan profiles of rice cell-derived rhGAA to identify the core-fucosylated glycans using UPLC and tandem mass spectrometry.
Collapse
Affiliation(s)
- Jihye Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Jonghye Do
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Hong-Yeol Choi
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, South Korea
| | - Sun-Dal Kim
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, South Korea
| | - Heajin Park
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Seungkwan You
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Wooseok Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Yeonjoo Jang
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Donghwi Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Junmyoung Lee
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Jongkwan Ha
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Minkyoo Ji
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea
| | - Dong-Il Kim
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, South Korea
| | - Ha Hyung Kim
- Biotherapeutics and Glycomics Laboratory, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea.
| |
Collapse
|
29
|
Rodriguez-Hernandez M, Triggiani D, Ivison F, Demurtas OC, Illiano E, Marino C, Franconi R, Massa S. Expression of a Functional Recombinant Human Glycogen Debranching Enzyme (hGDE) in N. benthamiana Plants and in Hairy Root Cultures. Protein Pept Lett 2020; 27:145-157. [PMID: 31622193 DOI: 10.2174/0929866526666191014154047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 05/14/2019] [Accepted: 08/02/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glycogen storage disease type III (GSDIII, Cori/Forbes disease) is a metabolic disorder due to the deficiency of the Glycogen Debranching Enzyme (GDE), a large monomeric protein (about 176 kDa) with two distinct enzymatic activities: 4-α-glucantransferase and amylo-α-1,6-glucosidase. Several mutations along the amylo-alpha-1,6-glucosidase,4-alphaglucanotransferase (Agl) gene are associated with loss of enzymatic activity. The unique treatment for GSDIII, at the moment, is based on diet. The potential of plants to manufacture exogenous engineered compounds for pharmaceutical purposes, from small to complex protein molecules such as vaccines, antibodies and other therapeutic/prophylactic entities, was shown by modern biotechnology through "Plant Molecular Farming". OBJECTIVE AND METHODS In an attempt to develop novel protein-based therapeutics for GSDIII, the Agl gene, encoding for the human GDE (hGDE) was engineered for expression as a histidinetagged GDE protein both in Nicotiana benthamiana plants by a transient expression approach, and in axenic hairy root in vitro cultures (HR) from Lycopersicum esculentum and Beta vulgaris. RESULTS In both plant-based expression formats, the hGDE protein accumulated in the soluble fraction of extracts. The plant-derived protein was purified by affinity chromatography in native conditions showing glycogen debranching activity. CONCLUSION These investigations will be useful for the design of a new generation of biopharmaceuticals based on recombinant GDE protein that might represent, in the future, a possible therapeutic option for GSDIII.
Collapse
Affiliation(s)
- Meilyn Rodriguez-Hernandez
- Center for Genetic Engineering and Biotechnology (CIGB), Direction of Agricultural Biotechnology, Havana,Cuba
| | - Doriana Triggiani
- Italian Glycogen Storage Disease Association (AIG) NPO, Assago, Milan, Italy
- Department of Sustainability (SSPT), Biomedical Technologies Laboratory, Italian National Agency for New Technologies, Energy and Sustainable Economic Development ENEA, Rome,Italy
| | - Fiona Ivison
- Department of Biochemistry, Manchester University NHS Foundation Trust, Manchester,United Kingdom
| | - Olivia C Demurtas
- Department of Sustainability (SSPT), Biotechnology Laboratory, ENEA, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome,Italy
| | - Elena Illiano
- Department of Sustainability (SSPT), Biomedical Technologies Laboratory, Italian National Agency for New Technologies, Energy and Sustainable Economic Development ENEA, Rome,Italy
| | - Carmela Marino
- Department of Sustainability (SSPT), Biomedical Technologies Laboratory, Italian National Agency for New Technologies, Energy and Sustainable Economic Development ENEA, Rome,Italy
| | - Rosella Franconi
- Department of Sustainability (SSPT), Biomedical Technologies Laboratory, Italian National Agency for New Technologies, Energy and Sustainable Economic Development ENEA, Rome,Italy
| | - Silvia Massa
- Department of Sustainability (SSPT), Biotechnology Laboratory, ENEA, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome,Italy
| |
Collapse
|
30
|
Clark M, Maselko M. Transgene Biocontainment Strategies for Molecular Farming. FRONTIERS IN PLANT SCIENCE 2020; 11:210. [PMID: 32194598 PMCID: PMC7063990 DOI: 10.3389/fpls.2020.00210] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/11/2020] [Indexed: 05/21/2023]
Abstract
Advances in plant synthetic biology promise to introduce novel agricultural products in the near future. 'Molecular farms' will include crops engineered to produce medications, vaccines, biofuels, industrial enzymes, and other high value compounds. These crops have the potential to reduce costs while dramatically increasing scales of synthesis and provide new economic opportunities to farmers. Current transgenic crops may be considered safe given their long-standing use, however, some applications of molecular farming may pose risks to human health and the environment. Unwanted gene flow from engineered crops could potentially contaminate the food supply, and affect wildlife. There is also potential for unwanted gene flow into engineered crops which may alter their ability to produce compounds of interest. Here, we briefly discuss the applications of molecular farming and explore the various genetic and physical methods that can be used for transgene biocontainment. As yet, no technology can be applied to all crop species, such that a combination of approaches may be necessary. Effective biocontainment is needed to enable large scale molecular farming.
Collapse
Affiliation(s)
- Michael Clark
- Applied Biosciences, Macquarie University, North Ryde, NSW, Australia
| | - Maciej Maselko
- Applied Biosciences, Macquarie University, North Ryde, NSW, Australia
- CSIRO Health and Biosecurity, Canberra, ACT, Australia
- CSIRO Synthetic Biology Future Science Platform, Brisbane, QLD, Australia
- *Correspondence: Maciej Maselko,
| |
Collapse
|
31
|
Tambuyzer E, Vandendriessche B, Austin CP, Brooks PJ, Larsson K, Miller Needleman KI, Valentine J, Davies K, Groft SC, Preti R, Oprea TI, Prunotto M. Therapies for rare diseases: therapeutic modalities, progress and challenges ahead. Nat Rev Drug Discov 2019; 19:93-111. [PMID: 31836861 DOI: 10.1038/s41573-019-0049-9] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/26/2022]
Abstract
Most rare diseases still lack approved treatments despite major advances in research providing the tools to understand their molecular basis, as well as legislation providing regulatory and economic incentives to catalyse the development of specific therapies. Addressing this translational gap is a multifaceted challenge, for which a key aspect is the selection of the optimal therapeutic modality for translating advances in rare disease knowledge into potential medicines, known as orphan drugs. With this in mind, we discuss here the technological basis and rare disease applicability of the main therapeutic modalities, including small molecules, monoclonal antibodies, protein replacement therapies, oligonucleotides and gene and cell therapies, as well as drug repurposing. For each modality, we consider its strengths and limitations as a platform for rare disease therapy development and describe clinical progress so far in developing drugs based on it. We also discuss selected overarching topics in the development of therapies for rare diseases, such as approval statistics, engagement of patients in the process, regulatory pathways and digital tools.
Collapse
Affiliation(s)
- Erik Tambuyzer
- BioPontis Alliance for Rare Diseases Foundation fup/son, Brussels, Belgium. .,BioPontis Alliance Rare Disease Foundation, Inc, Raleigh, NC, USA.
| | - Benjamin Vandendriessche
- Byteflies, Antwerp, Belgium.,Department of Electrical, Computer, and Systems Engineering (ECSE), Case Western Reserve University, Cleveland, OH, USA
| | - Christopher P Austin
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Philip J Brooks
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kristina Larsson
- Orphan Medicines Office, European Medicines Agency, Amsterdam, Netherlands
| | | | | | - Kay Davies
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Stephen C Groft
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Robert Preti
- Hitachi Chemical Regenerative Medicine Business Sector, Allendale, NJ, USA
| | - Tudor I Oprea
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico Albuquerque, Albuquerque, NM, USA.,UNM Comprehensive Cancer Center, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Marco Prunotto
- School of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
32
|
Quality Control and Downstream Processing of Therapeutic Enzymes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:55-80. [PMID: 31482494 DOI: 10.1007/978-981-13-7709-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Therapeutic enzymes are a commercially minor but clinically important area of biopharmaceuticals. An array of therapeutic enzymes has been developed for a variety of human diseases, including leukaemia and enzyme-deficiency diseases such as Gaucher's disease. Production and testing of therapeutic enzymes is strictly governed by regulatory bodies in each country around the world, and batch-to-batch consistency is crucially important. Manufacture of a batch starts with the fermentation or cell culture stage. After expression of the therapeutic enzyme in a cell culture bioreactor, robust and reproducible protein purification, or downstream processing (DSP) of the target product, is critical to ensuring safe delivery of these medicines. Modern processing technology, including the use of disposable processing equipment, has greatly improved the DSP development pathway in terms of robustness and speed to clinic. Once purified, the drug substance undergoes rigorous quality control (QC) testing according to current regulatory guidance, to enable release to the clinic and patient. QC testing is conducted to ensure the safety, purity, identity, potency and strength of the medicinal product, requiring multiple analytical methods that are rigorously validated and monitored for robust performance. Several case studies, including L-asparaginase and asfotase alfa, are discussed to illustrate the methods described herein.
Collapse
|
33
|
Daniell H, Kulis M, Herzog RW. Plant cell-made protein antigens for induction of Oral tolerance. Biotechnol Adv 2019; 37:107413. [PMID: 31251968 PMCID: PMC6842683 DOI: 10.1016/j.biotechadv.2019.06.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 05/21/2019] [Accepted: 06/24/2019] [Indexed: 12/15/2022]
Abstract
The gut associated lymphoid tissue has effective mechanisms in place to maintain tolerance to food antigens. These can be exploited to induce antigen-specific tolerance for the prevention and treatment of autoimmune diseases and severe allergies and to prevent serious immune responses in protein replacement therapies for genetic diseases. An oral tolerance approach for the prevention of peanut allergy in infants proved highly efficacious and advances in treatment of peanut allergy have brought forth an oral immunotherapy drug that is currently awaiting FDA approval. Several other protein antigens made in plant cells are in clinical development. Plant cell-made proteins are protected in the stomach from acids and enzymes after their oral delivery because of bioencapsulation within plant cell wall, but are released to the immune system upon digestion by gut microbes. Utilization of fusion protein technologies facilitates their delivery to the immune system, oral tolerance induction at low antigen doses, resulting in efficient induction of FoxP3+ and latency-associated peptide (LAP)+ regulatory T cells that express immune suppressive cytokines such as IL-10. LAP and IL-10 expression represent potential biomarkers for plant-based oral tolerance. Efficacy studies in hemophilia dogs support clinical development of oral delivery of bioencapsulated antigens to prevent anti-drug antibody formation. Production of clinical grade materials in cGMP facilities, stability of antigens in lyophilized plant cells for several years when stored at ambient temperature, efficacy of oral delivery of human doses in large animal models and lack of toxicity augur well for clinical advancement of this novel drug delivery concept.
Collapse
Affiliation(s)
- Henry Daniell
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Michael Kulis
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Roland W Herzog
- Department of Pediatrics, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
34
|
Jansing J, Sack M, Augustine SM, Fischer R, Bortesi L. CRISPR/Cas9-mediated knockout of six glycosyltransferase genes in Nicotiana benthamiana for the production of recombinant proteins lacking β-1,2-xylose and core α-1,3-fucose. PLANT BIOTECHNOLOGY JOURNAL 2019; 17:350-361. [PMID: 29969180 PMCID: PMC6335070 DOI: 10.1111/pbi.12981] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/11/2018] [Accepted: 06/25/2018] [Indexed: 05/19/2023]
Abstract
Plants offer fast, flexible and easily scalable alternative platforms for the production of pharmaceutical proteins, but differences between plant and mammalian N-linked glycans, including the presence of β-1,2-xylose and core α-1,3-fucose residues in plants, can affect the activity, potency and immunogenicity of plant-derived proteins. Nicotiana benthamiana is widely used for the transient expression of recombinant proteins so it is desirable to modify the endogenous N-glycosylation machinery to allow the synthesis of complex N-glycans lacking β-1,2-xylose and core α-1,3-fucose. Here, we used multiplex CRISPR/Cas9 genome editing to generate N. benthamiana production lines deficient in plant-specific α-1,3-fucosyltransferase and β-1,2-xylosyltransferase activity, reflecting the mutation of six different genes. We confirmed the functional gene knockouts by Sanger sequencing and mass spectrometry-based N-glycan analysis of endogenous proteins and the recombinant monoclonal antibody 2G12. Furthermore, we compared the CD64-binding affinity of 2G12 glycovariants produced in wild-type N. benthamiana, the newly generated FX-KO line, and Chinese hamster ovary (CHO) cells, confirming that the glyco-engineered antibody performed as well as its CHO-produced counterpart.
Collapse
Affiliation(s)
- Julia Jansing
- Department for Molecular BiotechnologyRWTH Aachen UniversityAachenGermany
| | - Markus Sack
- Department for Molecular BiotechnologyRWTH Aachen UniversityAachenGermany
| | | | - Rainer Fischer
- Department for Molecular BiotechnologyRWTH Aachen UniversityAachenGermany
- Present address:
Indiana Biosciences Research InstituteIndianapolisINUSA
- Present address:
Aachen‐Maastricht Institute for Biobased MaterialsMaastricht UniversityGeleenThe Netherlands
| | - Luisa Bortesi
- Department for Molecular BiotechnologyRWTH Aachen UniversityAachenGermany
- Present address:
Aachen‐Maastricht Institute for Biobased MaterialsMaastricht UniversityGeleenThe Netherlands
| |
Collapse
|
35
|
Buyel JF. Plant Molecular Farming - Integration and Exploitation of Side Streams to Achieve Sustainable Biomanufacturing. FRONTIERS IN PLANT SCIENCE 2019; 9:1893. [PMID: 30713542 PMCID: PMC6345721 DOI: 10.3389/fpls.2018.01893] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/06/2018] [Indexed: 05/22/2023]
Abstract
Plants have unique advantages over other systems such as mammalian cells for the production of valuable small molecules and proteins. The benefits cited most often include safety due to the absence of replicating human pathogens, simplicity because sterility is not required during production, scalability due to the potential for open-field cultivation with transgenic plants, and the speed of transient expression potentially providing gram quantities of product in less than 4 weeks. Initially there were also significant drawbacks, such as the need to clarify feed streams with a high particle burden and the large quantities of host cell proteins, but efficient clarification is now readily achieved. Several additional advantages have also emerged reflecting the fact that plants are essentially biodegradable, single-use bioreactors. This article will focus on the exploitation of this concept for the production of biopharmaceutical proteins, thus improving overall process economics. Specifically, we will discuss the single-use properties of plants, the sustainability of the production platform, and the commercial potential of different biomass side streams. We find that incorporating these side streams through rational process integration has the potential to more than double the revenue that can currently be achieved using plant-based production systems.
Collapse
Affiliation(s)
- Johannes F. Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
36
|
Zimran A, Dinur T, Revel-Vilk S, Akkerman EM, van Dussen L, Hollak CEM, Maayan H, Altarescu G, Chertkoff R, Maas M. Improvement in bone marrow infiltration in patients with type I Gaucher disease treated with taliglucerase alfa. J Inherit Metab Dis 2018; 41:1259-1265. [PMID: 30066229 PMCID: PMC6326976 DOI: 10.1007/s10545-018-0195-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 12/16/2022]
Abstract
Preliminary data suggest a positive effect of taliglucerase alfa on the bone marrow infiltration of Gaucher cells. In this investigator-initiated study, we report the impact of taliglucerase alfa on the bone marrow fat fraction (FF) in 26 patients assessed by quantitative chemical shift imaging (QCSI). Of 15 treatment-naïve patients (median age 48 [range 24-68] years), eight had baseline FF ≤ 0.3, six of those with a FF ≤ 0.23 ('bone at risk'). All significantly improved from a median baseline FF of 0.24 (0.15-0.32) to 1st year FF of 0.37 (0.25-0.54) and 2nd year FF of 0.42 (0.27-0.59) (p = 0.01). Among the 11 'switch-over' patients (median age 42 [range 33-69] years; median imiglucerase exposure 8 [range 1-17] years), eight had baseline FF ≤ 0.3, five of those with FF < 0.23. All, but one, significantly improved from a median baseline FF of 0.17 (0.08-0.28) to 1st year FF of 0.3 (0.05-0.34) and 2nd year FF of 0.34 (0.08-0.44) (p = 0.03). Two elderly female patients (age 43 and 58 years, with 17 years imiglucerase exposure) who remained at the same enzyme replacement therapy dose, increased from baseline FF of 0.13 and 0.19 to 0.26 at 1 year. Although the number of observations is small, we hypothesize that switching to taliglucerase may result in an improved bone marrow response. A larger study is needed to assess the early benefit of taliglucerase alfa in adult patients with type 1 Gaucher disease on the bone marrow compartment.
Collapse
Affiliation(s)
- Ari Zimran
- Gaucher Clinic, Shaare Zedek Medical Center, affiliated with Hebrew-University Medical School, Jerusalem, Israel
| | - Tama Dinur
- Gaucher Clinic, Shaare Zedek Medical Center, affiliated with Hebrew-University Medical School, Jerusalem, Israel
| | - Shoshana Revel-Vilk
- Gaucher Clinic, Shaare Zedek Medical Center, affiliated with Hebrew-University Medical School, Jerusalem, Israel.
| | | | | | | | - Hannah Maayan
- Gaucher Clinic, Shaare Zedek Medical Center, affiliated with Hebrew-University Medical School, Jerusalem, Israel
| | | | | | - Mario Maas
- Academic Medical Centre, Amsterdam, Netherlands
| |
Collapse
|
37
|
Hadi M, Swinburn P, Nalysnyk L, Hamed A, Mehta A. A health state utility valuation study to assess the impact of treatment mode of administration in Gaucher disease. Orphanet J Rare Dis 2018; 13:159. [PMID: 30201003 PMCID: PMC6131903 DOI: 10.1186/s13023-018-0903-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/29/2018] [Indexed: 01/05/2023] Open
Abstract
Background This study aimed to obtain UK societal-based utility values for health states related to treatment mode of administration using Gaucher disease as the background condition. Methods A review of relevant literature and expert clinical input informed the development of five health states characterising the impact of Gaucher disease and its management on patients’ lives. A base-state characterising the “controlled disease” was developed as well as four subsequent health states which varied in description of the method (intravenous versus oral) and frequency of treatment administration. Health state utilities were obtained using the time trade-off (TTO) method via face-to-face interviews with 100 members from the UK general population. Before the valuation exercise, participants provided informed consent, completed a demographic form and the EQ-5D, and ranked the health states from best to worst on a 0–100 visual analogue scale (VAS). Results Mean age of the participants (n = 100) was 35 years and 66% were female. Participants reported high EQ-5D VAS (86.1) and index scores (0.95) indicating very good health status. The “controlled disease” state had the highest mean TTO-derived utility value (0.89). There was only a marginal reduction in utility for the generic state for “Oral treatment” (0.85), while the reduction was more pronounced for the generic state for “Intravenous treatment” (0.73). Conclusions The findings suggest that the avoidance of the need for intravenous treatment administration is associated with a notable positive increase in health-related quality of life. Patient benefit arising from less invasive treatment could be an important consideration when undertaking economic evaluation of future therapies for Gaucher disease. Electronic supplementary material The online version of this article (10.1186/s13023-018-0903-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Luba Nalysnyk
- Sanofi Genzyme, 50 Binney Street, Cambridge, MA, 02142, USA.
| | - Alaa Hamed
- Sanofi Genzyme, 50 Binney Street, Cambridge, MA, 02142, USA
| | - Atul Mehta
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free Hospital and University College Medical School, London, UK
| |
Collapse
|
38
|
Oishi I, Yoshii K, Miyahara D, Tagami T. Efficient production of human interferon beta in the white of eggs from ovalbumin gene-targeted hens. Sci Rep 2018; 8:10203. [PMID: 29976933 PMCID: PMC6033876 DOI: 10.1038/s41598-018-28438-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022] Open
Abstract
Transgenic chickens could potentially serve as bioreactors for commercial production of recombinant proteins in egg white. Many transgenic chickens have been generated by randomly integrating viral vectors into their genomes, but transgene expression has proved insufficient and/or limited to the initial cohort. Herein, we demonstrate the feasibility of integrating human interferon beta (hIFN-β) into the chicken ovalbumin locus and producing hIFN-β in egg white. We knocked in hIFN-β into primordial germ cells using a CRISPR/Cas9 protocol and then generated germline chimeric roosters by cell transplantation into recipient embryos. Two generation-zero founder roosters produced hIFN-β knock-in offspring, and all knock-in female offspring produced abundant egg-white hIFN-β (~3.5 mg/ml). Although female offspring of the first generation were sterile, their male counterparts were fertile and produced a second generation of knock-in hens, for which egg-white hIFN-β production was comparable with that of the first generation. The hIFN-β bioactivity represented only ~5% of total egg-white hIFN-β, but unfolding and refolding of hIFN-β in the egg white fully recovered the bioactivity. These results suggest that transgene insertion at the chicken ovalbumin locus can result in abundant and stable expression of an exogenous protein deposited into egg white and should be amenable to industrial applications.
Collapse
Affiliation(s)
- Isao Oishi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, 1-8-31, Midorioka, Ikeda, Osaka, 563-8577, Japan.
| | - Kyoko Yoshii
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, 1-8-31, Midorioka, Ikeda, Osaka, 563-8577, Japan
| | - Daichi Miyahara
- Animal Breeding and Reproduction Research Division, National Agriculture and Food Research Organization, Institute of Livestock and Grassland Science, 2 Ikenodai, Tsukuba, Ibaraki, 305-0901, Japan
| | - Takahiro Tagami
- Animal Breeding and Reproduction Research Division, National Agriculture and Food Research Organization, Institute of Livestock and Grassland Science, 2 Ikenodai, Tsukuba, Ibaraki, 305-0901, Japan
| |
Collapse
|
39
|
Rozov SM, Permyakova NV, Deineko EV. Main Strategies of Plant Expression System Glycoengineering for Producing Humanized Recombinant Pharmaceutical Proteins. BIOCHEMISTRY (MOSCOW) 2018; 83:215-232. [PMID: 29625542 DOI: 10.1134/s0006297918030033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Most the pharmaceutical proteins are derived not from their natural sources, rather their recombinant analogs are synthesized in various expression systems. Plant expression systems, unlike mammalian cell cultures, combine simplicity and low cost of procaryotic systems and the ability for posttranslational modifications inherent in eucaryotes. More than 50% of all human proteins and more than 40% of the currently used pharmaceutical proteins are glycosylated, that is, they are glycoproteins, and their biological activity, pharmacodynamics, and immunogenicity depend on the correct glycosylation pattern. This review examines in detail the similarities and differences between N- and O-glycosylation in plant and mammalian cells, as well as the effect of plant glycans on the activity, pharmacokinetics, immunity, and intensity of biosynthesis of pharmaceutical proteins. The main current strategies of glycoengineering of plant expression systems aimed at obtaining fully humanized proteins for pharmaceutical application are summarized.
Collapse
Affiliation(s)
- S M Rozov
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | | | | |
Collapse
|
40
|
Plants as sources of natural and recombinant anti-cancer agents. Biotechnol Adv 2018; 36:506-520. [DOI: 10.1016/j.biotechadv.2018.02.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/23/2018] [Accepted: 02/01/2018] [Indexed: 02/07/2023]
|
41
|
Zimran A, Wajnrajch M, Hernandez B, Pastores GM. Taliglucerase alfa: safety and efficacy across 6 clinical studies in adults and children with Gaucher disease. Orphanet J Rare Dis 2018; 13:36. [PMID: 29471850 PMCID: PMC5824466 DOI: 10.1186/s13023-018-0776-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/15/2018] [Indexed: 11/10/2022] Open
Abstract
Taliglucerase alfa is an enzyme replacement therapy (ERT) approved for treatment of adult and paediatric patients with Type 1 Gaucher disease (GD) in several countries and the first plant cell-expressed recombinant therapeutic protein approved by the US Food and Drug Administration for humans. Here, we review the findings across six key taliglucerase alfa clinical studies. A total of 33 treatment-naïve adult patients were randomized to taliglucerase alfa 30 U/kg or 60 U/kg in a 9-month, multicentre, randomized, double-blind, parallel-group, dose-comparison pivotal study, after which eligible patients continued into two consecutive extension studies; 17 treatment-naïve adult patients completed 5 total years of treatment with taliglucerase alfa. In the only ERT study focused on exclusively paediatric patients with GD, 11 treatment-naïve children were randomized to taliglucerase alfa 30 U/kg or 60 U/kg in a 12-month, multicentre, double-blind study; nine completed 3 total years of treatment in a dedicated paediatric extension study. The effect of switching patients from imiglucerase to taliglucerase alfa was also investigated in a separate 9-month study that included 26 adults and five children; 10 adults completed a total of 3 years and two children completed a total of 2.75 years of taliglucerase alfa treatment in the extension studies. All studies evaluated safety and spleen volume, liver volume, platelet count, haemoglobin concentration, and biomarkers as measures of efficacy. Detailed results from baseline through the end of these studies are presented. Taliglucerase alfa was well tolerated, and adverse events were generally mild/moderate in severity and transient. Treatment with taliglucerase alfa resulted in improvements (treatment-naïve patients) or stability (patients switched from imiglucerase) in visceral, haematologic, and biomarker parameters. Together, this comprehensive data set supports the treatment of adult and paediatric patients with GD who are naïve to ERT or who have previously been treated with imiglucerase.
Collapse
Affiliation(s)
- Ari Zimran
- Gaucher Clinic, Shaare Zedek Medical Center, Hebrew University and Hadassah Medical School, 12 Bayit Street, P.O. Box 3235, 91031, Jerusalem, Israel.
| | | | | | - Gregory M Pastores
- University College Dublin and the National Centre for Inherited Metabolic Disorders, Mater Misericordiae University Hospital, Dublin, Ireland
| |
Collapse
|
42
|
Farelli JD, Asrani KH, Isaacs C, deBear JS, Stahley MR, Shah A, Lasaro MA, Cheng CJ, Subramanian RR. Leveraging Rational Protein Engineering to Improve mRNA Therapeutics. Nucleic Acid Ther 2018; 28:74-85. [PMID: 29437538 DOI: 10.1089/nat.2017.0697] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Messenger RNA (mRNA) is a promising new class of therapeutics that has potential for treatment of diseases in fields such as immunology, oncology, vaccines, and inborn errors of metabolism. mRNA therapy has several advantages over DNA-based gene therapy, including the lack of the need for nuclear import and transcription, as well as limited possibility of genomic integration. One drawback of mRNA therapy, especially in cases such as metabolic disorders where repeated dosing will be necessary, is the relatively short in vivo half-life of mRNA (∼6-12 h). We hypothesize that protein engineering designed to improve translation, yielding longer-lasting protein, or modifications that would increase enzymatic activity would be helpful in alleviating this issue. In this study, we present two examples where sequence engineering improved the expression and duration, as well as enzymatic activity of target proteins in vitro. We then confirmed these findings in wild-type mice. This work shows that rational engineering of proteins can lead to improved therapies in vivo.
Collapse
Affiliation(s)
- Jeremiah D Farelli
- 1 Discovery Research, Alexion Pharmaceuticals, Inc. , Cambridge, Massachusetts
| | - Kirtika H Asrani
- 1 Discovery Research, Alexion Pharmaceuticals, Inc. , Cambridge, Massachusetts
| | - Cleo Isaacs
- 1 Discovery Research, Alexion Pharmaceuticals, Inc. , Cambridge, Massachusetts
| | - Joanna S deBear
- 2 Nucleic Acid Technology, Alexion Pharmaceuticals, Inc. , New Haven, Connecticut
| | - Mary R Stahley
- 2 Nucleic Acid Technology, Alexion Pharmaceuticals, Inc. , New Haven, Connecticut
| | - Anumeha Shah
- 3 Preclinical Pharmacology, Alexion Pharmaceuticals, Inc. , New Haven, Connecticut
| | - Melissa A Lasaro
- 3 Preclinical Pharmacology, Alexion Pharmaceuticals, Inc. , New Haven, Connecticut
| | - Christopher J Cheng
- 2 Nucleic Acid Technology, Alexion Pharmaceuticals, Inc. , New Haven, Connecticut
| | | |
Collapse
|
43
|
Glyco-Engineering of Plant-Based Expression Systems. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 175:137-166. [PMID: 30069741 DOI: 10.1007/10_2018_76] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Most secreted proteins in eukaryotes are glycosylated, and after a number of common biosynthesis steps the glycan structures mature in a species-dependent manner. Therefore, human therapeutic proteins produced in plants often carry plant-like rather than human-like glycans, which can affect protein stability, biological function, and immunogenicity. The glyco-engineering of plant-based expression systems began as a strategy to eliminate plant-like glycans and produce human proteins with authentic or at least compatible glycan structures. The precise replication of human glycans is challenging, owing to the absence of a pathway in plants for the synthesis of sialylated proteins and the necessary precursors, but this can now be achieved by the coordinated expression of multiple human enzymes. Although the research community has focused on the removal of plant glycans and their replacement with human counterparts, the presence of plant glycans on proteins can also provide benefits, such as boosting the immunogenicity of some vaccines, facilitating the interaction between therapeutic proteins and their receptors, and increasing the efficacy of antibody effector functions. Graphical Abstract Typical structures of native mammalian and plant glycans with symbols indicating sugar residues identified by their short form and single-letter codes. Both glycans contain fucose, albeit with different linkages.
Collapse
|
44
|
Engineering of Yeast Glycoprotein Expression. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 175:93-135. [DOI: 10.1007/10_2018_69] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Gupta P, Pastores GM. Spotlight on taliglucerase alfa in the treatment of pediatric patients with type 1 Gaucher disease. PEDIATRIC HEALTH MEDICINE AND THERAPEUTICS 2017; 8:73-81. [PMID: 29388611 PMCID: PMC5774592 DOI: 10.2147/phmt.s93634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Gaucher disease (GD) is a heritable storage disorder caused by functional defects of the lysosomal acid β-glucosidase and the accumulation of glucosylceramide within macrophages, resulting in multiple organ dysfunction. There are three commercially available enzyme replacement therapy (ERT) products for the treatment of GD type 1 (GD1): imiglucerase, velaglucerase alfa, and taliglucerase alfa. Imiglucerase and velaglucerase alfa are produced in different mammalian cell systems; imiglucerase requires postproduction deglycosylation to expose terminal α-mannose residues, which are required for mannose receptor-mediated uptake by target macrophages. These steps are critical to the success of ERT for the treatment of visceral and hematologic manifestations of GD. Taliglucerase alfa is the first US Food and Drug Administration-approved plant-cell-expressed recombinant human protein, using carrot root cell cultures. Furthermore, it does not require postproduction glycosidic modifications. It is indicated for treatment of adults with GD1 in the US, Israel, Australia, Canada, Chile, Brazil, and other countries, and it is additionally approved for the treatment of pediatric patients in the US, Australia, and Canada and for the treatment of hematologic manifestations in pediatric patients with Type 3 GD in Canada and other countries. Our review focuses on the role of taliglucerase alfa in the pediatric population. A literature search through PubMed (from 1995 up till November 2016) of English language articles was performed with the following terms: Gaucher disease, lysosomal storage disease, taliglucerase. Secondary and tertiary references were obtained by reviewing related articles as well as the website www.Clinicaltrials.gov. It has been demonstrated that taliglucerase alfa is efficacious, with a well-established safety profile in pediatric, ERT-naïve patients with symptomatic GD1, as well as for those patients previously treated with imiglucerase.
Collapse
Affiliation(s)
- Punita Gupta
- Division of Genetics, Department of Pediatrics, St. Joseph's Children's Hospital, Paterson, New Jersey, USA
| | - Gregory M Pastores
- National Center for Inherited Metabolic Disorders, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| |
Collapse
|
46
|
Jung JW, Huy NX, Kim HB, Kim NS, Van Giap D, Yang MS. Production of recombinant human acid α-glucosidase with high-mannose glycans in gnt1 rice for the treatment of Pompe disease. J Biotechnol 2017; 249:42-50. [PMID: 28363873 DOI: 10.1016/j.jbiotec.2017.03.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/27/2017] [Accepted: 03/27/2017] [Indexed: 10/19/2022]
Abstract
Lysosomal storage diseases are a group of inherited metabolic disorders. Patients are treated with enzyme replacement therapy (ERT), in which the replacement enzymes are required to carry terminal mannose or mannose 6-phosphate residues to allow efficient uptake into target cells and tissues. N-acetylglucosaminyltransferase-I (GnTI) mediates N-glycosylation in the cis cisternae of the Golgi apparatus by adding N-acetylglucosamine to the exposed terminal mannose residue of core N-glycan structures for further processing. Mutant rice lacking GnTI produces only high mannosylated glycoproteins. In this study, we introduced a gene encoding recombinant human acid α-glucosidase (rhGAA), which is used in ERT for Pompe disease, into gnt1 rice callus by particle bombardment. Integration of the target gene into the genome of the gnt1 rice line and its mRNA expression were confirmed by PCR and Northern blot, respectively. Western blot analysis was performed to confirm secretion of the target proteins into the culture media. Using an indirect enzyme linked immunosorbent assay, we determined the maximum expression of rhGAA to be approximately 45mg/L, 13days after induction. To assay the enzymatic activity and determine the N-glycan profile of rhGAA, we purified the protein using a 6×histidine tag. The in vitro α-glucosidase activity of rhGAA from gnt1 rice callus (gnt1-GAA) was 3.092U/mg, similar to the activity of the Chinese hamster ovary cell-derived GAA (3.154U/mg). N-glycan analysis revealed the presence of high-mannose N-glycans on gnt1-GAA. In addition, the production of high-mannose GAA using gnt1 rice calli as an expression host was characterized, which may aid the future development of therapeutic enzymes for the treatment of Pompe disease.
Collapse
Affiliation(s)
- Jae-Wan Jung
- Department of Molecular Biology, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea; Department of Bioactive Material Science, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea
| | - Nguyen-Xuan Huy
- Department of Molecular Biology, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea; Biology Department, Hue University of Education, 34 Le Loi, Hue, Viet Nam
| | - Hyo-Boon Kim
- Department of Molecular Biology, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea
| | - Nan-Sun Kim
- Department of Molecular Biology, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea
| | - Do Van Giap
- Department of Bioactive Material Science, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea
| | - Moon-Sik Yang
- Department of Molecular Biology, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea; Department of Bioactive Material Science, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea; Research Center of Bioactive Materials, Chonbuk National University, 664-14 Dukjindong, Jeonju, Jeollabuk-do 561-756, Republic of Korea.
| |
Collapse
|
47
|
Wong-Arce A, González-Ortega O, Rosales-Mendoza S. Plant-Made Vaccines in the Fight Against Cancer. Trends Biotechnol 2017; 35:241-256. [DOI: 10.1016/j.tibtech.2016.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 11/21/2016] [Accepted: 12/07/2016] [Indexed: 12/25/2022]
|
48
|
Lomonossoff GP, D'Aoust MA. Plant-produced biopharmaceuticals: A case of technical developments driving clinical deployment. Science 2017; 353:1237-40. [PMID: 27634524 DOI: 10.1126/science.aaf6638] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ability to express heterologous proteins in plants has led to the concept of using plants as "bioreactors" or "biofactories" for the production of pharmaceutical proteins. Although initial studies were promising, the pathway to commercialization and deployment in a clinical setting has proven to be a somewhat rocky road. This Review examines the technical developments that have led to the current increase in interest in the use of plants for the production of pharmaceutical proteins, particularly in the context of clinical trials.
Collapse
Affiliation(s)
- George P Lomonossoff
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK.
| | - Marc-André D'Aoust
- Medicago, 1020 Route de l'Église, Bureau 600, Quebec City, Quebec G1V 3V9, Canada
| |
Collapse
|
49
|
Lagassé HAD, Alexaki A, Simhadri VL, Katagiri NH, Jankowski W, Sauna ZE, Kimchi-Sarfaty C. Recent advances in (therapeutic protein) drug development. F1000Res 2017; 6:113. [PMID: 28232867 PMCID: PMC5302153 DOI: 10.12688/f1000research.9970.1] [Citation(s) in RCA: 297] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2017] [Indexed: 01/11/2023] Open
Abstract
Therapeutic protein drugs are an important class of medicines serving patients most in need of novel therapies. Recently approved recombinant protein therapeutics have been developed to treat a wide variety of clinical indications, including cancers, autoimmunity/inflammation, exposure to infectious agents, and genetic disorders. The latest advances in protein-engineering technologies have allowed drug developers and manufacturers to fine-tune and exploit desirable functional characteristics of proteins of interest while maintaining (and in some cases enhancing) product safety or efficacy or both. In this review, we highlight the emerging trends and approaches in protein drug development by using examples of therapeutic proteins approved by the U.S. Food and Drug Administration over the previous five years (2011–2016, namely January 1, 2011, through August 31, 2016).
Collapse
Affiliation(s)
- H A Daniel Lagassé
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Aikaterini Alexaki
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Vijaya L Simhadri
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Nobuko H Katagiri
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Wojciech Jankowski
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Zuben E Sauna
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Chava Kimchi-Sarfaty
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
50
|
Márquez-Escobar VA. Current developments and prospects on human metapneumovirus vaccines. Expert Rev Vaccines 2017; 16:419-431. [PMID: 28116910 DOI: 10.1080/14760584.2017.1283223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Human metapneumovirus (hMPV) has become one of the major pathogens causing acute respiratory infections (ARI) mainly affecting young children, immunocompromised patients, and the elderly. Currently there are no licensed vaccines against this virus. Areas covered: Since the discovery of hMPV in 2001, many groups have focused on developing vaccines against this pathogen. This review presents the outcomes and perspectives derived from preclinical studies performed in cell cultures and animals as well as the only candidate that has reached evaluation in a clinical trial. Limitations of the current vaccine candidates are discussed and perspectives for the development of plant-based vaccines are analyzed. Expert commentary: Several hMPV vaccine candidates are under development with the potential to progress into clinical trials. In parallel, the molecular farming field offers new opportunities to generate innovative vaccines that will offer several advantages in the fight against hMPV.
Collapse
Affiliation(s)
- Verónica Araceli Márquez-Escobar
- a Facultad de Ciencias Químicas , Universidad Autónoma de San Luis Potosí , Av. Dr. Manuel Nava 6, San Luis Potosí 78210 , SLP , Mexico
| |
Collapse
|