1
|
Mayer V, Steiner F, Jungbauer A, Pereira Aguilar P. Highly pure measles virus generated by combination of salt-active nuclease treatment and heparin affinity chromatography. J Chromatogr A 2024; 1738:465470. [PMID: 39488125 DOI: 10.1016/j.chroma.2024.465470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
Highly purified virus preparations are essential for accurate activity and potency determination. This requires simple and efficient purification methods, especially in the early stages of research and development. While heparin affinity chromatography has been already successfully used for the purification of several enveloped viruses and virus-like particles, we extended its use to purification of very sensitive measles virus. The performance of heparin and heparin-like affinity chromatography was evaluated for the purification of recombinant measles virus, a large and labile enveloped virus used as vaccine or cancer therapy. Since DNA, particularly in the form of chromatin is a critical impurity in enveloped virus preparations, the effect of integration of an endonuclease (Benzonase® or M-SAN) treatment prior to chromatography was also investigated. Both, Capto™ DeVirS (heparin-like) and Capto™ Heparin were able to capture measles viruses directly from clarified cell culture supernatant. Despite capturing 100 % of infectious measles virus, low recovery (8 %) was observed for Capto™ DeVirS. For Capto™ Heparin recoveries up to 85 % were observed. The combination of M-SAN with Capto™ Heparin enabled the production of highly purified measles virus with a yield of 62 % and a final purity of 10.2 ng dsDNA per dose (1 × 105), outperforming the processes without endonuclease treatment with a yield of 18 %, and a purity of 66.7 ng dsDNA/dose or using Benzonase® with a yield of 38 % and a purity of 21.2 ng dsDNA/dose. As the developed method is simple and scalable it could also be integrated in a downstream process train for measles virus manufacturing.
Collapse
Affiliation(s)
- Viktoria Mayer
- Austrian Centre of Industrial Biotechnology, Vienna, Austria; Institute of Bioprocess Science and Engineering (IBSE), BOKU University, Vienna, Austria
| | - Florian Steiner
- Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Alois Jungbauer
- Austrian Centre of Industrial Biotechnology, Vienna, Austria; Institute of Bioprocess Science and Engineering (IBSE), BOKU University, Vienna, Austria.
| | - Patricia Pereira Aguilar
- Austrian Centre of Industrial Biotechnology, Vienna, Austria; Institute of Bioprocess Science and Engineering (IBSE), BOKU University, Vienna, Austria
| |
Collapse
|
2
|
Stephens AD, Wilkinson T. Discovery of Therapeutic Antibodies Targeting Complex Multi-Spanning Membrane Proteins. BioDrugs 2024; 38:769-794. [PMID: 39453540 PMCID: PMC11530565 DOI: 10.1007/s40259-024-00682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Complex integral membrane proteins, which are embedded in the cell surface lipid bilayer by multiple transmembrane spanning polypeptides, encompass families of proteins that are important target classes for drug discovery. These protein families include G protein-coupled receptors, ion channels, transporters, enzymes, and adhesion molecules. The high specificity of monoclonal antibodies and the ability to engineer their properties offers a significant opportunity to selectively bind these target proteins, allowing direct modulation of pharmacology or enabling other mechanisms of action such as cell killing. Isolation of antibodies that bind these types of membrane proteins and exhibit the desired pharmacological function has, however, remained challenging due to technical issues in preparing membrane protein antigens suitable for enabling and driving antibody drug discovery strategies. In this article, we review progress and emerging themes in defining discovery strategies for a generation of antibodies that target these complex membrane protein antigens. We also comment on how this field may develop with the emerging implementation of computational techniques, artificial intelligence, and machine learning.
Collapse
Affiliation(s)
- Amberley D Stephens
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK
| | - Trevor Wilkinson
- Department of Biologics Engineering, Oncology R&D, The Discovery Centre, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0AA, UK.
| |
Collapse
|
3
|
Guzman Ruiz L, Zollner AM, Hoxie I, Küchler J, Hausjell C, Mesurado T, Krammer F, Jungbauer A, Pereira Aguilar P, Klausberger M, Grabherr R. Enhancing NA immunogenicity through novel VLP designs. Vaccine 2024; 42:126270. [PMID: 39197219 DOI: 10.1016/j.vaccine.2024.126270] [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: 06/27/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Current influenza virus vaccines poorly display key neuraminidase (NA) epitopes and do not robustly induce NA-reactive antibodies; instead, they focus on the induction of hemagglutinin (HA)-reactive antibodies. Next-generation influenza vaccines should be optimized in order to activate NA-reactive B cells and to induce a broadly cross-reactive and protective antibody response. We aimed at enhancing the immunogenicity of the NA on vaccines by two strategies: (i) modifying the HA:NA ratio of the vaccine preparation and (ii) exposing epitopes on the lateral surface or beneath the head of the NA by extending the NA stalk. The H1N1 glycoproteins from the influenza virus A/California/04/2009 strain were displayed on human immunodeficiency virus 1 (HIV-1) gag-based virus-like particles (VLP). Using the baculovirus insect cell expression system, we biased the quantity of surface glycoproteins employing two different promoters, the very late baculovirus p10 promoter and the early and late gp64 promoter. This led to a 1:1 to 2:1 HA:NA ratio, which was approximately double or triple the amount of NA as present on the wild-type influenza A virus (HA:NA ratio 3:1 to 5:1). Furthermore, by insertion of 15 amino acids from the A-New York/61/2012 strain (NY12) which prolongates the NA stalk (NA long stalk; NA-LS), we intended to improve the accessibility of the NA. Six different types of VLPs were produced and purified using a platform downstream process based on Capto-Core 700™ followed by Capto-Heparin™ affinity chromatography combined with ultracentrifugation. These VLPs were then tested in a mouse model. Robust titers of antibodies that inhibit the neuraminidase activity were elicited even after vaccination with two low doses (0.3 μg) of the H1N1 VLPs without compromising the anti-HA responses. In conclusion, our results demonstrate the feasibility of the two developed strategies to retain HA immunogenicity and improve NA immunogenicity as a future influenza vaccine candidate.
Collapse
Affiliation(s)
- Leticia Guzman Ruiz
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria; University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Alexander M Zollner
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Irene Hoxie
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, Gustave L. Levy Place, 10029-5674 New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jan Küchler
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Christina Hausjell
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria
| | - Tomas Mesurado
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria
| | - Florian Krammer
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, Gustave L. Levy Place, 10029-5674 New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Alois Jungbauer
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria
| | - Patricia Pereira Aguilar
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Bioprocess Science and Engineering (IBSE), Muthgasse 18, 1190 Vienna, Austria; acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria
| | - Miriam Klausberger
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria
| | - Reingard Grabherr
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Biotechnology, Institute of Molecular Biotechnology (IMBT), Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
4
|
Shastry S, Barbieri E, Minzoni A, Chu W, Johnson S, Stoops M, Pancorbo J, Gilleskie G, Ritola K, Crapanzano MS, Daniele MA, Menegatti S. Serotype-agnostic affinity purification of adeno-associated virus (AAV) via peptide-functionalized chromatographic resins. J Chromatogr A 2024; 1734:465320. [PMID: 39217737 DOI: 10.1016/j.chroma.2024.465320] [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: 07/08/2024] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Adeno-associated viruses (AAVs) have emerged as a prominent family of vectors for gene delivery, providing therapeutic options to diseases once deemed incurable. At the same time, they necessitate efficient and affordable purification methods that can be platformed to serve all AAV serotypes. Current chromatographic tools, while affording high product purity, fail to bind certain serotypes, provide limited yield and lifetime, and impose harsh elution conditions that can compromise the vector's activity and safety. Addressing these challenges, this work demonstrates the application of new peptide ligands as the first serotype-agnostic technology for AAV purification by affinity chromatography. Our study reveals a pH-dependent affinity interaction: AAV2, AAV3, AAV6, AAV9, and AAVrh.10 are effectively captured at neutral pH, while binding AAV1, AAV5, AAV7, and AAV8 is stronger in a slightly acidic environment. The elution of bound AAVs was achieved using magnesium chloride at neutral pH for all serotypes, consistently affording capsid yields above 50% and genome yields above 80%, together with a >100-fold reduction in host cell proteins and nucleic acids. In particular, peptide ligand A10 exhibited remarkable binding capacity (> 1014 vp per mL of resin) and purification performance for all AAV serotypes, demonstrating broad applicability for gene therapy manufacturing. Finally, this work introduces novel alkaline-stable variants of A10 and demonstrates their use as the first affinity ligands capable of performing multiple cycles of AAV2, AAV8, and AAV9 purification with intermediate caustic cleaning without loss of capacity or product quality. Collectively, these results demonstrate the promise of this technology to further the impact and affordability of gene therapy.
Collapse
Affiliation(s)
- Shriarjun Shastry
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27606, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
| | - Eduardo Barbieri
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27606, USA; LigaTrap Technologies LLC, Raleigh, NC 27606, USA
| | - Arianna Minzoni
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27606, USA
| | - Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27606, USA
| | - Stephanie Johnson
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
| | - Mark Stoops
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
| | - Jennifer Pancorbo
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
| | - Gary Gilleskie
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
| | - Kimberly Ritola
- Neuroscience Center, Brain Initiative Neurotools Vector Core, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 911 Oval Dr, Raleigh, NC 27695, USA
| | | | - Michael A Daniele
- North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 911 Oval Dr, Raleigh, NC 27695, USA; Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, 911 Oval Drive, Raleigh, NC 27695, USA
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27606, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 911 Oval Dr, Raleigh, NC 27695, USA; LigaTrap Technologies LLC, Raleigh, NC 27606, USA.
| |
Collapse
|
5
|
Valentic A, Hubbuch J. Effective removal of host cell-derived nucleic acids bound to hepatitis B core antigen virus-like particles by heparin chromatography. Front Bioeng Biotechnol 2024; 12:1475918. [PMID: 39431243 PMCID: PMC11487522 DOI: 10.3389/fbioe.2024.1475918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
Virus-like particles (VLPs) show considerable potential for a wide array of therapeutic applications, spanning from vaccines targeting infectious diseases to applications in cancer immunotherapy and drug delivery. In the context of hepatitis B core antigen (HBcAg) VLPs, a promising candidate for gene delivery approaches, the naturally occurring nucleic acid (NA) binding region is commonly utilized for effective binding of various types of therapeutic nucleic acids (NAther). During formation of the HBcAg VLPs, host cell-derived nucleic acids (NAhc) might be associated to the NA binding region, and are thus encapsulated into the VLPs. Following a VLP harvest, the NAhc need to be removed effectively before loading the VLP with NAther. Various techniques reported in literature for this NAhc removal, including enzymatic treatments, alkaline treatment, and lithium chloride precipitation, lack quantitative evidence of sufficient NAhc removal accompanied by a subsequent high VLP protein recovery. In this study, we present a novel heparin chromatography-based process for effective NAhc removal from HBcAg VLPs. Six HBcAg VLP constructs with varying lengths of the NA binding region and diverse NAhc loadings were subjected to evaluation. Process performance was thoroughly examined through NAhc removal and VLP protein recovery analyses. Hereby, reversed phase chromatography combined with UV/Vis spectroscopy, as well as silica spin column-based chromatography coupled with dye-based fluorescence assay were employed. Additionally, alternative process variants, comprising sulfate chromatography and additional nuclease treatments, were investigated. Comparative analyses were conducted with LiCl precipitation and alkaline treatment procedures to ascertain the efficacy of the newly developed chromatography-based methods. Results revealed the superior performance of the heparin chromatography procedure in achieving high NAhc removal and concurrent VLP protein recovery. Furthermore, nuanced relationships between NA binding region length and NAhc removal efficiency were elucidated. Hereby, the construct Cp157 surpassed the other constructs in the heparin process by demonstrating high NAhc removal and VLP protein recovery. Among the other process variants minimal performance variations were observed for the selected constructs Cp157 and Cp183. However, the heparin chromatography-based process consistently outperformed other methods, underscoring its superiority in NAhc removal and VLP protein recovery.
Collapse
Affiliation(s)
| | - Jürgen Hubbuch
- Institute of Process Engineering in Life Sciences – Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
6
|
Bobbili MR, Görgens A, Yan Y, Vogt S, Gupta D, Corso G, Barbaria S, Patrioli C, Weilner S, Pultar M, Jacak J, Hackl M, Schosserer M, Grillari R, Kjems J, Andaloussi SEL, Grillari J. Snorkel-tag based affinity chromatography for recombinant extracellular vesicle purification. J Extracell Vesicles 2024; 13:e12523. [PMID: 39400515 PMCID: PMC11472238 DOI: 10.1002/jev2.12523] [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: 04/30/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid nanoparticles and play an important role in cell-cell communications, making them potential therapeutic agents and allowing to engineer for targeted drug delivery. The expanding applications of EVs in next generation medicine is still limited by existing tools for scaling standardized EV production, single EV tracing and analytics, and thus provide only a snapshot of tissue-specific EV cargo information. Here, we present the Snorkel-tag, for which we have genetically fused the EV surface marker protein CD81, to a series of tags with an additional transmembrane domain to be displayed on the EV surface, resembling a snorkel. This system enables the affinity purification of EVs from complex matrices in a non-destructive form while maintaining EV characteristics in terms of surface protein profiles, associated miRNA patterns and uptake into a model cell line. Therefore, we consider the Snorkel-tag to be a widely applicable tool in EV research, allowing for efficient preparation of EV standards and reference materials, or dissecting EVs with different surface markers when fusing to other tetraspanins in vitro or in vivo.
Collapse
Affiliation(s)
- Madhusudhan Reddy Bobbili
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue Regeneration
| | - André Görgens
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Institute for Transfusion Medicine, University Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Yan Yan
- Department of Molecular Biology and Genetics, Centre for Cellular Signal Patterns (CellPat), Interdisciplinary Nanoscience Centre (iNANO)Aarhus UniversityAarhus CDenmark
- Omiics ApSAarhus NDenmark
| | - Stefan Vogt
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Dhanu Gupta
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Institute of Developmental and Regenerative MedicineUniversity of Oxford, IMS‐Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, HeadingtonOxfordUnited Kingdom
- Department of PaediatricsUniversity of Oxford, South Parks RoadOxfordUnited Kingdom
| | - Giulia Corso
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Evercyte GmbHViennaAustria
| | - Samir Barbaria
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Carolina Patrioli
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Sylvia Weilner
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | | | - Jaroslaw Jacak
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- School of Medical Engineering and Applied Social ScienceUniversity of Applied Sciences Upper AustriaLinzAustria
| | - Matthias Hackl
- Austrian Cluster for Tissue Regeneration
- TAmiRNATAmiRNA GmbHViennaAustria
| | - Markus Schosserer
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Austrian Cluster for Tissue Regeneration
- Institute of Medical GeneticsCenter for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Regina Grillari
- Austrian Cluster for Tissue Regeneration
- Evercyte GmbHViennaAustria
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Centre for Cellular Signal Patterns (CellPat), Interdisciplinary Nanoscience Centre (iNANO)Aarhus UniversityAarhus CDenmark
| | - Samir EL Andaloussi
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
| | - Johannes Grillari
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue Regeneration
| |
Collapse
|
7
|
Sripada SA, Barbieri E, Shastry S, Wuestenhagen E, Aldinger A, Rammo O, Schulte MM, Daniele M, Menegatti S. Multiangle Light Scattering as a Lentivirus Purification Process Analytical Technology. Anal Chem 2024; 96:9593-9600. [PMID: 38804040 DOI: 10.1021/acs.analchem.4c01209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The limited biomolecular and functional stability of lentiviral vectors (LVVs) for cell therapy poses the need for analytical tools that can monitor their titers and activity throughout the various steps of expression and purification. In this study, we describe a rapid (25 min) and reproducible (coefficient of variance ∼0.5-2%) method that leverages size exclusion chromatography coupled with multiangle light scattering detection (SEC-MALS) to determine size, purity, and particle count of LVVs purified from bioreactor harvests. The SEC-MALS data were corroborated by orthogonal methods, namely, dynamic light scattering (DLS) and transmission electron microscopy. The method was also evaluated for robustness in the range of 2.78 × 105-2.67 × 107 particles per sample. Notably, MALS-based particle counts correlated with the titer of infectious LVVs measured via transduction assays (R2 = 0.77). Using a combination of SEC-MALS and DLS, we discerned the effects of purification parameters on LVV quality, such as the separation between heterogeneous LV, which can facilitate critical decision-making in the biomanufacturing of gene and cell therapies.
Collapse
Affiliation(s)
- Sobhana A Sripada
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Raleigh, North Carolina 27606, United States
- NC-VVIRAL, NC State University, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
| | - Eduardo Barbieri
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Raleigh, North Carolina 27606, United States
- LigaTrap Technologies LLC, 1791 Varsity Drive, Suite #150, Raleigh, North Carolina 27606, United States
| | - Shriarjun Shastry
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Raleigh, North Carolina 27606, United States
- Biomanufacturing Training and Education Center, NC State University, 850 Oval Dr, Raleigh, North Carolina 27606, United States
- NC-VVIRAL, NC State University, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
| | | | | | | | | | - Michael Daniele
- Department of Electrical and Computer Engineering, NC State University, 890 Oval Dr, Raleigh, North Carolina 27606, United States
- Joint Department of Biomedical Engineering, NC State University and UNC Chapel Hill, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
- NC-VVIRAL, NC State University, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Raleigh, North Carolina 27606, United States
- LigaTrap Technologies LLC, 1791 Varsity Drive, Suite #150, Raleigh, North Carolina 27606, United States
- Biomanufacturing Training and Education Center, NC State University, 850 Oval Dr, Raleigh, North Carolina 27606, United States
- NC-VVIRAL, NC State University, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
| |
Collapse
|
8
|
Guzman Ruiz L, Zollner AM, Hoxie I, Arcalis E, Krammer F, Klausberger M, Jungbauer A, Grabherr R. Increased efficacy of influenza virus vaccine candidate through display of recombinant neuraminidase on virus like particles. Front Immunol 2024; 15:1425842. [PMID: 38915410 PMCID: PMC11194364 DOI: 10.3389/fimmu.2024.1425842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 06/26/2024] Open
Abstract
Vaccination against influenza virus can reduce the risk of influenza by 40% to 60%, they rely on the production of neutralizing antibodies specific to influenza hemagglutinin (HA) ignoring the neuraminidase (NA) as an important surface target. Vaccination with standardized NA concentration may offer broader and longer-lasting protection against influenza infection. In this regard, we aimed to compare the potency of a NA displayed on the surface of a VLP with a soluble NA. The baculovirus expression system (BEVS) and the novel virus-free Tnms42 insect cell line were used to express N2 NA on gag-based VLPs. To produce VLP immunogens with high levels of purity and concentration, a two-step chromatography purification process combined with ultracentrifugation was used. In a prime/boost vaccination scheme, mice vaccinated with 1 µg of the N2-VLPs were protected from mortality, while mice receiving the same dose of unadjuvanted NA in soluble form succumbed to the lethal infection. Moreover, NA inhibition assays and NA-ELISAs of pre-boost and pre-challenge sera confirm that the VLP preparation induced higher levels of NA-specific antibodies outperforming the soluble unadjuvanted NA.
Collapse
Affiliation(s)
- Leticia Guzman Ruiz
- Institute of Molecular Biotechnology (IMBT), Department of Biotechnology (DBT), University of Natural Resources and Life Sciences Vienna (BOKU), Vienna, Austria
- Institute of Bioprocess Science and Engineering (IBSE), Department of Biotechnology (DBT), University of Natural Resources and Life Sciences Vienna (BOKU), Vienna, Austria
| | - Alexander M. Zollner
- Institute of Bioprocess Science and Engineering (IBSE), Department of Biotechnology (DBT), University of Natural Resources and Life Sciences Vienna (BOKU), Vienna, Austria
| | - Irene Hoxie
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Elsa Arcalis
- Institute of Plant Biotechnology and Cell Biology (IPBT), Department of Applied Genetics and Cell Biology (DAGZ), University of Natural Resources and Life Sciences Vienna (BOKU), Vienna, Austria
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Miriam Klausberger
- Institute of Molecular Biotechnology (IMBT), Department of Biotechnology (DBT), University of Natural Resources and Life Sciences Vienna (BOKU), Vienna, Austria
| | - Alois Jungbauer
- Institute of Bioprocess Science and Engineering (IBSE), Department of Biotechnology (DBT), University of Natural Resources and Life Sciences Vienna (BOKU), Vienna, Austria
- Austrian Centre of Industrial Biotechnology (acib), Vienna, Austria
| | - Reingard Grabherr
- Institute of Molecular Biotechnology (IMBT), Department of Biotechnology (DBT), University of Natural Resources and Life Sciences Vienna (BOKU), Vienna, Austria
| |
Collapse
|
9
|
Achleitner L, Winter M, Aguilar PP, Lingg N, Jungbauer A, Klausberger M, Satzer P. Robust and resource-efficient production process suitable for large-scale production of baculovirus through high cell density seed train and optimized infection strategy. N Biotechnol 2024; 80:46-55. [PMID: 38302001 DOI: 10.1016/j.nbt.2024.01.002] [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: 10/24/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
The aim of this study was the development of a scalable production process for high titer (108 pfu/mL and above) recombinant baculovirus stocks with low cell line-derived impurities for the production of virus-like particles (VLP). To achieve this, we developed a high cell density (HCD) culture for low footprint cell proliferation, compared different infection strategies at multiplicity of infection (MOI) 0.05 and 0.005, different infection strategies and validated generally applicable harvest criteria of cell viability ≤ 80%. We also investigated online measurable parameters to observe the baculovirus production. The infection strategy employing a very low virus inoculum of MOI 0.005 and a 1:2 dilution with fresh medium one day after infection proved to be the most resource efficient. There, we achieved higher cell-specific titers and lower host cell protein concentrations at harvest than other tested infection strategies with the same MOI, while saving half of the virus stock for infecting the culture compared to other tested infection strategies. HCD culture by daily medium exchange was confirmed as suitable for seed train propagation, infection, and baculovirus production, equally efficient as the conventionally propagated seed train. Online measurable parameters for cell concentration and average cell diameter were found to be effective in monitoring the production process. The study concluded that a more efficient VLP production process in large scale can be achieved using this virus stock production strategy, which could also be extended to produce other proteins or extracellular vesicles with the baculovirus expression system.
Collapse
Affiliation(s)
- Lena Achleitner
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Martina Winter
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Patricia Pereira Aguilar
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Nico Lingg
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Alois Jungbauer
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Miriam Klausberger
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Peter Satzer
- acib - Austrian Centre of Industrial Biotechnology, Muthgasse 11, 1190 Vienna, Austria; Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna (BOKU), Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
10
|
Ma J, Tian Z, Shi Q, Dong X, Sun Y. Affinity chromatography for virus-like particle manufacturing: Challenges, solutions, and perspectives. J Chromatogr A 2024; 1721:464851. [PMID: 38574547 DOI: 10.1016/j.chroma.2024.464851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
The increasing medical application of virus-like particles (VLPs), notably vaccines and viral vectors, has increased the demand for commercial VLP production. However, VLP manufacturing has not yet reached the efficiency level achieved for recombinant protein therapeutics, especially in downstream processing. This review provides a comprehensive analysis of the challenges associated with affinity chromatography for VLP purification with respect to the diversity and complexity of VLPs and the associated upstream and downstream processes. The use of engineered affinity ligands and matrices for affinity chromatography is first discussed. Although several representative affinity ligands are currently available for VLP purification, most of them have difficulty in balancing ligand universality, ligand selectivity and mild operation conditions. Then, phage display technology and computer-assisted design are discussed as efficient methods for the rapid discovery of high-affinity peptide ligands. Finally, the VLP purification by affinity chromatography is analyzed. The process is significantly influenced by virus size and variation, ligand type and chromatographic mode. To address the updated regulatory requirements and epidemic outbreaks, technical innovations in affinity chromatography and process intensification and standardization in VLP purification should be promoted to achieve rapid process development and highly efficient VLP manufacturing, and emphasis is given to the discovery of universal ligands, applications of gigaporous matrices and platform technology. It is expected that the information in this review can provide a better understanding of the affinity chromatography methods available for VLP purification and offer useful guidance for the development of affinity chromatography for VLP manufacturing in the decades to come.
Collapse
Affiliation(s)
- Jing Ma
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Zengquan Tian
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Qinghong Shi
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China.
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China.
| |
Collapse
|
11
|
LeBarre JP, Chu W, Altern SH, Kocot AJ, Bhandari D, Barbieri E, Sly J, Crapanzano M, Cramer SM, Phillips M, Roush D, Carbonell R, Boi C, Menegatti S. Mixed-mode size-exclusion silica resin for polishing human antibodies in flow-through mode. J Chromatogr A 2024; 1720:464772. [PMID: 38452560 DOI: 10.1016/j.chroma.2024.464772] [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: 09/22/2023] [Revised: 02/07/2024] [Accepted: 02/25/2024] [Indexed: 03/09/2024]
Abstract
The polishing step in the downstream processing of therapeutic antibodies removes residual impurities from Protein A eluates. Among the various classes of impurities, antibody fragments are especially challenging to remove due to the broad biomolecular diversity generated by a multitude of fragmentation patterns. The current approach to fragment removal relies on ion exchange or mixed-mode adsorbents operated in bind-and-gradient-elution mode. However, fragments that bear strong similarity to the intact product or whose biophysical features deviate from the ensemble average can elude these adsorbents, and the lack of a chromatographic technology enabling robust antibody polishing is recognized as a major gap in downstream bioprocessing. Responding to this challenge, this study introduces size-exclusion mixed-mode (SEMM) silica resins as a novel chromatographic adsorbent for the capture of antibody fragments irrespective of their biomolecular features. The pore diameter of the silica beads features a narrow distribution and is selected to exclude monomeric antibodies, while allowing their fragments to access the pores where they are captured by the mixed-mode ligands. The static and dynamic binding capacity of the adsorbent ranged respectively between 30-45 and 25-33 gs of antibody fragments per liter of resin. Selected SEMM-silica resins also demonstrated the ability to capture antibody aggregates, which adsorb on the outer layer of the beads. Optimization of the SEMM-silica design and operation conditions - namely, pore size (10 nm) and ligand composition (quaternary amine and alkyl chain) as well as the linear velocity (100 cm/h), ionic strength (5.7 mS/cm), and pH (7) of the mobile phase - afforded a significant reduction of both fragments and aggregates, resulting into a final antibody yield up to 80% and monomeric purity above 97%.
Collapse
Affiliation(s)
- Jacob P LeBarre
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Scott H Altern
- The Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Andrew J Kocot
- The Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Dipendra Bhandari
- LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA
| | - Eduardo Barbieri
- LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA
| | - Jae Sly
- LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA
| | - Michael Crapanzano
- LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA
| | - Steven M Cramer
- The Howard P. Isermann Department of Chemical and Biological Engineering and the Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | | | - David Roush
- Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, Roush Biopharma Panacea, 20 Squire Terrace, Colts Neck, NJ, 07033, USA
| | - Ruben Carbonell
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
| | - Cristiana Boi
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA; Department of Civil, Chemical Environmental and Materials Engineering, University of Bologna, Via Terracini 28, 40131, Bologna, Italy
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; LigaTrap Technologies, Raleigh, 1791 Varsity Dr, Raleigh, NC, 27606, USA; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA.
| |
Collapse
|
12
|
Maeda K, Goto S, Miura K, Saito K, Morita E. The incorporation of extracellular vesicle markers varies among vesicles with distinct surface charges. J Biochem 2024; 175:299-312. [PMID: 38030385 DOI: 10.1093/jb/mvad097] [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: 08/24/2023] [Accepted: 11/11/2023] [Indexed: 12/01/2023] Open
Abstract
Extracellular vesicles (EVs) are important mediators of intercellular communication. However, the methods available for distinguishing the heterogeneity of secreted EVs and isolating and purifying them are limited. This study introduced a HiBiT-tag to detect various EV markers, including CD63, CD9, Epidermal Growth Factor Receptor (EGFR), Flotilin1, and Syndecan-1, and investigated whether these marker-containing vesicles were capable of binding to differently charged column carriers. Four column carriers, Diethylaminoethyl (DEAE), Capto Adhere, Blue and Heparin, showed affinity for CD63 containing EVs, but their elution patterns varied. Furthermore, we observed that the elution patterns of the EV markers differed among vesicles with distinct surface charges when a DEAE column was used. This suggests that the incorporation of EV markers varied between these vesicles. The markers showed different subcellular localizations, indicating that the site of vesicle formation may contribute to the production of vesicles with varying charges and marker incorporation. These findings may have implications for the development of methods to purify homogeneous EVs, which could be useful in EV-mediated drug delivery systems.
Collapse
Grants
- 20333747, 19fk0108168h0001, 20he0622012h0001, 22fk0108527s0101 AMED
- 23790503, 26460555, 16H01188, 17H06413, 20 K21874, 22 K18378, 22H02873, 22H00553 JSPS KAKENHI
- Japan, and the Takeda Medical Research Foundation
- JPMJCR17H4 JST CREST
Collapse
Affiliation(s)
- Koki Maeda
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosakishi, Aomori 036-8561, Japan
- Division of Biomolecular Function, Bioresources Science, United Graduate School of Agricultural Sciences, Iwate University, 3 Bunkyo-cho, Hirosakishi, Aomori 036-8561, Japan
| | - Simon Goto
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosakishi, Aomori 036-8561, Japan
| | - Koya Miura
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosakishi, Aomori 036-8561, Japan
| | - Koki Saito
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosakishi, Aomori 036-8561, Japan
- Division of Biomolecular Function, Bioresources Science, United Graduate School of Agricultural Sciences, Iwate University, 3 Bunkyo-cho, Hirosakishi, Aomori 036-8561, Japan
| | - Eiji Morita
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosakishi, Aomori 036-8561, Japan
- Division of Biomolecular Function, Bioresources Science, United Graduate School of Agricultural Sciences, Iwate University, 3 Bunkyo-cho, Hirosakishi, Aomori 036-8561, Japan
| |
Collapse
|
13
|
Bonner SE, van de Wakker SI, Phillips W, Willms E, Sluijter JPG, Hill AF, Wood MJA, Vader P. Scalable purification of extracellular vesicles with high yield and purity using multimodal flowthrough chromatography. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e138. [PMID: 38939900 PMCID: PMC11080796 DOI: 10.1002/jex2.138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are cell derived membranous nanoparticles. EVs are important mediators of cell-cell communication via the transfer of bioactive content and as such they are being investigated for disease diagnostics as biomarkers and for potential therapeutic cargo delivery to recipient cells. However, existing methods for isolating EVs from biological samples suffer from challenges related to co-isolation of unwanted materials such as proteins, nucleic acids, and lipoproteins. In the pursuit of improved EV isolation techniques, we introduce multimodal flowthrough chromatography (MFC) as a scalable alternative to size exclusion chromatography (SEC). The use of MFC offers significant advantages for purifying EVs, resulting in enhanced yields and increased purity with respect to protein and nucleic acid co-isolates from conditioned 3D cell culture media. Compared to SEC, significantly higher EV yields with similar purity and preserved functionality were also obtained with MFC in 2D cell cultures. Additionally, MFC yielded EVs from serum with comparable purity to SEC and similar apolipoprotein B content. Overall, MFC presents an advancement in EV purification yielding EVs with high recovery, purity, and functionality, and offers an accessible improvement to researchers currently employing SEC.
Collapse
Affiliation(s)
| | - Simonides I. van de Wakker
- Department of Experimental CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - William Phillips
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| | - Eduard Willms
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVictoriaAustralia
| | - Joost P. G. Sluijter
- Department of Experimental CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityMelbourneVictoriaAustralia
| | | | - Pieter Vader
- Department of Experimental CardiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
14
|
Kilgore R, Minzoni A, Shastry S, Smith W, Barbieri E, Wu Y, LeBarre JP, Chu W, O'Brien J, Menegatti S. The downstream bioprocess toolbox for therapeutic viral vectors. J Chromatogr A 2023; 1709:464337. [PMID: 37722177 DOI: 10.1016/j.chroma.2023.464337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/20/2023]
Abstract
Viral vectors are poised to acquire a prominent position in modern medicine and biotechnology owing to their role as delivery agents for gene therapies, oncolytic agents, vaccine platforms, and a gateway to engineer cell therapies as well as plants and animals for sustainable agriculture. The success of viral vectors will critically depend on the availability of flexible and affordable biomanufacturing strategies that can meet the growing demand by clinics and biotech companies worldwide. In this context, a key role will be played by downstream process technology: while initially adapted from protein purification media, the purification toolbox for viral vectors is currently undergoing a rapid expansion to fit the unique biomolecular characteristics of these products. Innovation efforts are articulated on two fronts, namely (i) the discovery of affinity ligands that target adeno-associated virus, lentivirus, adenovirus, etc.; (ii) the development of adsorbents with innovative morphologies, such as membranes and 3D printed monoliths, that fit the size of viral vectors. Complementing these efforts are the design of novel process layouts that capitalize on novel ligands and adsorbents to ensure high yield and purity of the product while safeguarding its therapeutic efficacy and safety; and a growing panel of analytical methods that monitor the complex array of critical quality attributes of viral vectors and correlate them to the purification strategies. To help explore this complex and evolving environment, this study presents a comprehensive overview of the downstream bioprocess toolbox for viral vectors established in the last decade, and discusses present efforts and future directions contributing to the success of this promising class of biological medicines.
Collapse
Affiliation(s)
- Ryan Kilgore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States.
| | - Arianna Minzoni
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Shriarjun Shastry
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695, United States
| | - Will Smith
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Eduardo Barbieri
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Yuxuan Wu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Jacob P LeBarre
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Juliana O'Brien
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, United States
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States; Biomanufacturing Training and Education Center (BTEC), North Carolina State University, Raleigh, NC 27695, United States; North Carolina Viral Vector Initiative in Research and Learning, North Carolina State University, Raleigh, NC 27695, United States
| |
Collapse
|
15
|
Saari H, Pusa R, Marttila H, Yliperttula M, Laitinen S. Development of tandem cation exchange chromatography for high purity extracellular vesicle isolation: The effect of ligand steric availability. J Chromatogr A 2023; 1707:464293. [PMID: 37579702 DOI: 10.1016/j.chroma.2023.464293] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/16/2023]
Abstract
Purification of extracellular vesicles for research and therapeutic applications requires updated methodology to address the limitations of traditional ultracentrifugation and other size-based separation techniques. Their downfalls include induced extracellular vesicle aggregation, low yields, poor scalability and one-dimensionality of the separation process, as the size or sedimentation speed of extracellular vesicles is often the only selection criterion. Ion exchange chromatography is a promising alternative or supplementary method candidate, as it offers a different approach for extracellular vesicle separation, which is surface charge. For now, mostly anion exchange chromatography has been evaluated for extracellular vesicle purification, as it successfully relies on the strongly negative surface charge of extracellular vesicles. However, as extracellular vesicles are very complex in their structure, also cation exchange chromatography could be applicable, due to individual cationic domains on the extracellular vesicle surface. Here, we compare anion exchange chromatography to different types of cation exchange chromatography for the purification of platelet extracellular vesicle samples also containing plasma-derived impurities. We found that the choice of resin structure used for cation exchange chromatography is critical for binding platelet extracellular vesicles, as a conventional-type cation exchanger was found to only capture and elute less than 20% of extracellular vesicles. With the tentacle-type resin, it was possible to obtain comparable platelet extracellular vesicle yields (over 90%) with cation exchange chromatography compared to anion exchange chromatography, as well as superior purity, especially when it was combined to conventional cation exchange resin.
Collapse
Affiliation(s)
- Heikki Saari
- Finnish Red Cross Blood Service. Härkälenkki 13, 01730 Vantaa, Finland.
| | - Reetta Pusa
- Finnish Red Cross Blood Service. Härkälenkki 13, 01730 Vantaa, Finland
| | - Heli Marttila
- Molecular and Integrative Biosciences Research Programme, Biological and Environmental Sciences, University of Helsinki, Viikinkaari 9, 00790 Helsinki, Finland
| | - Marjo Yliperttula
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, 00790 Helsinki, Finland
| | - Saara Laitinen
- Finnish Red Cross Blood Service. Härkälenkki 13, 01730 Vantaa, Finland
| |
Collapse
|
16
|
Lothert K, Harsy YMJ, Endres P, Müller E, Wolff MW. Evaluation of restricted access media for the purification of cell culture-derived Orf viruses. Eng Life Sci 2023; 23:e2300009. [PMID: 37664009 PMCID: PMC10472920 DOI: 10.1002/elsc.202300009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 09/05/2023] Open
Abstract
Recently, multimodal chromatography using restricted access media (RAM) for the purification of nanoparticles, such as viruses has regained increasing attention. These chromatography resins combine size exclusion on the particle shell and adsorptive interaction within the core. Accordingly, smaller process-related impurities, for example, DNA and proteins, can be retained, while larger product viruses can pass unhindered. We evaluated a range of currently available RAM, differing in the shells' pore cut-off and the core chemistry, for the purification of a cell culture-derived clarified model virus, namely the Orf virus (ORFV). We examined impurity depletion and product recovery as relevant criteria for the evaluation of column performance, as well as scale-up robustness and regeneration potential for evaluating a multiple use application. The results indicate that some columns, for example, the Capto Core, enable both a high DNA and protein removal, while others, for example, the Monomix Core 60 (MC60), are more suitable for DNA depletion. Furthermore, column regeneration is facilitated by using columns with larger shell pores (5000 vs. 700 kDa) and weaker binding interactions (anion exchange vs. multimodal). According to these findings, the choice of RAM resins should be selected according to the respective feed sample composition and the planned number of application cycles.
Collapse
Affiliation(s)
- Keven Lothert
- Institute of Bioprocess Engineering and Pharmaceutical TechnologyUniversity of Applied Sciences Mittelhessen (THM)GiessenGermany
| | - Yasmina M. J. Harsy
- Institute of Bioprocess Engineering and Pharmaceutical TechnologyUniversity of Applied Sciences Mittelhessen (THM)GiessenGermany
| | - Patrick Endres
- Tosoh Bioscience GmbH, Separations Business Unit ‐ EuropeGriesheimGermany
| | - Egbert Müller
- Tosoh Bioscience GmbH, Separations Business Unit ‐ EuropeGriesheimGermany
| | - Michael W. Wolff
- Institute of Bioprocess Engineering and Pharmaceutical TechnologyUniversity of Applied Sciences Mittelhessen (THM)GiessenGermany
| |
Collapse
|
17
|
Lorenzo E, Miranda L, Gòdia F, Cervera L. Downstream process design for Gag HIV-1 based virus-like particles. Biotechnol Bioeng 2023; 120:2672-2684. [PMID: 37148527 DOI: 10.1002/bit.28419] [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: 11/24/2022] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 05/08/2023]
Abstract
Virus-like particles-based vaccines have been gaining interest in recent years. The manufacturing of these particles includes their production by cell culture followed by their purification to meet the requirements of its final use. The presence of host cell extracellular vesicles represents a challenge for better virus-like particles purification, because both share similar characteristics which hinders their separation. The present study aims to compare some of the most used downstream processing technologies for capture and purification of virus-like particles. Four steps of the purification process were studied, including a clarification step by depth filtration and filtration, an intermediate step by tangential flow filtration or multimodal chromatography, a capture step by ion exchange, heparin affinity and hydrophobic interaction chromatography and finally, a polishing step by size exclusion chromatography. In each step, the yields were evaluated by percentage of recovery of the particles of interest, purity, and elimination of main contaminants. Finally, a complete purification train was implemented using the best results obtained in each step. A final concentration of 1.40 × 1010 virus-like particles (VLPs)/mL with a purity of 64% after the polishing step was achieved, with host cell DNA and protein levels complaining with regulatory standards, and an overall recovery of 38%. This work has resulted in the development of a purification process for HIV-1 Gag-eGFP virus-like particles suitable for scale-up.
Collapse
Affiliation(s)
- Elianet Lorenzo
- Departament d'Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Laia Miranda
- Departament d'Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, Bellaterra, Spain
- University College London, London, UK
| | - Francesc Gòdia
- Departament d'Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Laura Cervera
- Departament d'Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
18
|
Lothert K, Wolff MW. Affinity and Pseudo-Affinity Membrane Chromatography for Viral Vector and Vaccine Purifications: A Review. MEMBRANES 2023; 13:770. [PMID: 37755191 PMCID: PMC10537005 DOI: 10.3390/membranes13090770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023]
Abstract
Several chromatographic approaches have been established over the last decades for the production of pharmaceutically relevant viruses. Due to the large size of these products compared to other biopharmaceuticals, e.g., proteins, convective flow media have proven to be superior to bead-based resins in terms of process productivity and column capacity. One representative of such convective flow materials is membranes, which can be modified to suit the particular operating principle and are also suitable for economical single-use applications. Among the different membrane variants, affinity surfaces allow for the most selective separation of the target molecule from other components in the feed solution, especially from host cell-derived DNA and proteins. A successful membrane affinity chromatography, however, requires the identification and implementation of ligands, which can be applied economically while at the same time being stable during the process and non-toxic in the case of any leaching. This review summarizes the current evaluation of membrane-based affinity purifications for viruses and virus-like particles, including traditional resin and monolith approaches and the advantages of membrane applications. An overview of potential affinity ligands is given, as well as considerations of suitable affinity platform technologies, e.g., for different virus serotypes, including a description of processes using pseudo-affinity matrices, such as sulfated cellulose membrane adsorbers.
Collapse
Affiliation(s)
| | - Michael W. Wolff
- Institute of Bioprocess Engineering and Pharmaceutical Technology, Department Life Science Engineering, University of Applied Sciences Mittelhessen (THM), 35390 Giessen, Germany
| |
Collapse
|
19
|
Mayer V, Frank AC, Preinsperger S, Csar P, Steppert P, Jungbauer A, Pereira Aguilar P. Removal of chromatin by salt-tolerant endonucleases for production of recombinant measles virus. Biotechnol Prog 2023; 39:e3342. [PMID: 36974026 DOI: 10.1002/btpr.3342] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/27/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Host cell DNA is a critical impurity in downstream processing of enveloped viruses. Especially, DNA in the form of chromatin is often neglected. Endonuclease treatment is an almost mandatory step in manufacturing of viral vaccines. In order to find the optimal performer, four different endonucleases, two of them salt tolerant, were evaluated in downstream processing of recombinant measles virus. Endonuclease treatment was performed under optimal temperature conditions after clarification and before the purification by flow-through chromatography with a core shell chromatography medium: Capto™ Core 700. Virus infectivity was measured by TCID50. DNA and histone presence in process and purified samples was determined using PicoGreen™ assay and Western blot analysis using an anti-histone antibody, respectively. All tested endonucleases allowed the reduction of DNA content improving product purity. The salt-tolerant endonucleases SAN and M-SAN were more efficient in the removal of chromatin compared with the non-salt-tolerant endonucleases Benzonase® and DENARASE®. Removal of chromatin using M-SAN was also possible without the addition of extra salt to the cell culture supernatant. The combination of the endonuclease treatment, using salt-tolerant endonucleases with flow-through chromatography, using core-shell particles, resulted in high purity and purification efficiency. This strategy has all features for a platform downstream process of recombinant measles virus and beyond.
Collapse
Affiliation(s)
- Viktoria Mayer
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Anna-Carina Frank
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Shirin Preinsperger
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Patrick Csar
- Themis Bioscience GmbH (A Subsidiary of Merck & Co., Inc, Kenilworth, NJ, USA), Vienna, Austria
| | - Petra Steppert
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Alois Jungbauer
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Patricia Pereira Aguilar
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
20
|
Zhou Y, Yuan R, Cone AS, Shifflett KW, Arias GF, Peng A, Chambers MG, McNamara RP, Willcox S, Landis JT, Pan Y, Griffith J, Dittmer DP. Large-scale heparin-based bind-and-elute chromatography identifies two biologically distinct populations of extracellular vesicles. J Extracell Vesicles 2023; 12:e12327. [PMID: 37272197 PMCID: PMC10240191 DOI: 10.1002/jev2.12327] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/10/2023] [Accepted: 04/20/2023] [Indexed: 06/06/2023] Open
Abstract
Purifying extracellular vesicles (EVs) has been challenging because EVs are heterogeneous in cargo yet share similar sizes and densities. Most surface marker-based affinity separation methods are limited to research or diagnostic scales. We report that heparin chromatography can separate purified EVs into two distinct subpopulations as ascertained by MS/MS: a non-heparin-binding (NHB) fraction that contains classical EV markers such as tetraspanins and a heparin-binding (HB) fraction enriched in fibronectins and histones. Both fractions were similarly fusogenic but induced different transcriptional responses in endothelial cells. While EVs that were purified by conventional, non-affinity methods alone induced ERK1/2 phosphorylation and Ki67, the NHB fraction did not. This result suggests heparin chromatography as an additional novel fractionation step that is inherently scalable, does not lead to loss of material, and separates inflammatory and pyrogenic EVs from unreactive EVs, which will improve clinical applications.
Collapse
Affiliation(s)
- Yijun Zhou
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Runjie Yuan
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Allaura S. Cone
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Kyle W. Shifflett
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Gabriel F. Arias
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Biochemistry and BiophysicsThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Alice Peng
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Meredith G. Chambers
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Ryan P. McNamara
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Smaranda Willcox
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Justin T. Landis
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Yue Pan
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of BiostatisticsThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Jack Griffith
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Biochemistry and BiophysicsThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Dirk P. Dittmer
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Department of Microbiology and ImmunologyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
21
|
Hillebrandt N, Hubbuch J. Size-selective downstream processing of virus particles and non-enveloped virus-like particles. Front Bioeng Biotechnol 2023; 11:1192050. [PMID: 37304136 PMCID: PMC10248422 DOI: 10.3389/fbioe.2023.1192050] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Non-enveloped virus-like particles (VLPs) are versatile protein nanoparticles with great potential for biopharmaceutical applications. However, conventional protein downstream processing (DSP) and platform processes are often not easily applicable due to the large size of VLPs and virus particles (VPs) in general. The application of size-selective separation techniques offers to exploit the size difference between VPs and common host-cell impurities. Moreover, size-selective separation techniques offer the potential for wide applicability across different VPs. In this work, basic principles and applications of size-selective separation techniques are reviewed to highlight their potential in DSP of VPs. Finally, specific DSP steps for non-enveloped VLPs and their subunits are reviewed as well as the potential applications and benefits of size-selective separation techniques are shown.
Collapse
Affiliation(s)
| | - Jürgen Hubbuch
- Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
22
|
De Figueiredo I, Bartenlian B, Van der Rest G, Pallandre A, Halgand F. Proteomics Methodologies: The Search of Protein Biomarkers Using Microfluidic Systems Coupled to Mass Spectrometry. Proteomes 2023; 11:proteomes11020019. [PMID: 37218924 DOI: 10.3390/proteomes11020019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/29/2023] [Accepted: 05/07/2023] [Indexed: 05/24/2023] Open
Abstract
Protein biomarkers have been the subject of intensive studies as a target for disease diagnostics and monitoring. Indeed, biomarkers have been extensively used for personalized medicine. In biological samples, these biomarkers are most often present in low concentrations masked by a biologically complex proteome (e.g., blood) making their detection difficult. This complexity is further increased by the needs to detect proteoforms and proteome complexity such as the dynamic range of compound concentrations. The development of techniques that simultaneously pre-concentrate and identify low-abundance biomarkers in these proteomes constitutes an avant-garde approach to the early detection of pathologies. Chromatographic-based methods are widely used for protein separation, but these methods are not adapted for biomarker discovery, as they require complex sample handling due to the low biomarker concentration. Therefore, microfluidics devices have emerged as a technology to overcome these shortcomings. In terms of detection, mass spectrometry (MS) is the standard analytical tool given its high sensitivity and specificity. However, for MS, the biomarker must be introduced as pure as possible in order to avoid chemical noise and improve sensitivity. As a result, microfluidics coupled with MS has become increasingly popular in the field of biomarker discovery. This review will show the different approaches to protein enrichment using miniaturized devices and the importance of their coupling with MS.
Collapse
Affiliation(s)
- Isabel De Figueiredo
- Institut de Chimie Physique, Université Paris Saclay, Avenue Jean Perrin, F91400 Orsay, France
| | - Bernard Bartenlian
- Centre des Nanosciences et Nanotechnologies, Université Paris Saclay, 10 Boulevard Thomas Gobert, F91120 Palaiseau, France
| | - Guillaume Van der Rest
- Institut de Chimie Physique, Université Paris Saclay, Avenue Jean Perrin, F91400 Orsay, France
| | - Antoine Pallandre
- Institut de Chimie Physique, Université Paris Saclay, Avenue Jean Perrin, F91400 Orsay, France
| | - Frédéric Halgand
- Institut de Chimie Physique, Université Paris Saclay, Avenue Jean Perrin, F91400 Orsay, France
| |
Collapse
|
23
|
Gritti F. Absorption and escape kinetics of spherical biomolecules from fully porous particles utilized in size exclusion chromatography. J Chromatogr A 2023; 1701:464050. [PMID: 37216849 DOI: 10.1016/j.chroma.2023.464050] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023]
Abstract
The increasing demand for the characterization of large biomolecules such as monoclonal antibodies, double-stranded deoxyribonucleic acid (dsDNA), and virus-like particles (VLPs) is raising fundamental questions pertaining to their absorption (ingress) and escape (egress) kinetics from fully porous particles. The exact expression of their concentration profiles is derived as a function of time and radial position across a single sub-3 μm Bridge-Ethylene-Hybrid (BEHTM) Particle present in size exclusion chromatography (SEC) columns. The boundary condition at the external surface area of the particle is a rectangular concentration profile mimicking the passage of the chromatographic zone. Four different BEH Particles were considered in the calculations depending on the molecular size of the analyte: 2.0 μm 100 Å BEH Particles for small molecules, 2.0 μm 200 Å BEH Particles for monoclonal antibodies, 2.0 μm 300 Å BEH Particles for dsDNA (100 base pairs), and 2.5 μm 900 Å BEH Particles for virus-like particles (VLPs). The calculated concentration profiles of small molecules and monoclonal antibodies confirm that all BEH Particles present in the column reach quasi-instantaneously thermodynamic equilibrium with the bulk mobile phase during the passage of the chromatographic band. This is no longer the case for larger biomolecules such as dsDNA or VLPs, especially when the SEC particle is located near the column inlet and for high velocities. The kinetics of biomolecule egress is slower than its kinetics of ingress leading to pronounced peak tailing. The mean concentration of the largest biomolecules in the SEC particles remains always smaller than the maximum bulk concentration. This persistent and transient intra-particle diffusion regime has direct implications on the theoretical expressions of the observed retention factors and plate heights. Classical theories of chromatography assume uniform spatial distribution of the analyte in the particle volume: this hypothesis is not verified for the largest biomolecules. These results imply that non-porous particles or monolithic structures are the most promising stationary phases for the separation and purification of the largest biomolecules in life science.
Collapse
Affiliation(s)
- Fabrice Gritti
- Waters Corporation, Instrument/Core Research/Fundamentals, 34 Maple Street, Milford, MA 01757, USA.
| |
Collapse
|
24
|
Mazurov D, Ramadan L, Kruglova N. Packaging and Uncoating of CRISPR/Cas Ribonucleoproteins for Efficient Gene Editing with Viral and Non-Viral Extracellular Nanoparticles. Viruses 2023; 15:v15030690. [PMID: 36992399 PMCID: PMC10056905 DOI: 10.3390/v15030690] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Rapid progress in gene editing based on clustered regularly interspaced short palindromic repeats/CRISPR-associated protein (CRISPR/Cas) has revolutionized functional genomic studies and genetic disease correction. While numerous gene editing applications have been easily adapted by experimental science, the clinical utility of CRISPR/Cas remains very limited due to difficulty in delivery to primary cells and possible off-target effects. The use of CRISPR in the form of a ribonucleoprotein (RNP) complex substantially reduces the time of DNA exposure to the effector nuclease and minimizes its off-target activity. The traditional electroporation and lipofection methods lack the cell-type specificity of RNP delivery, can be toxic for cells, and are less efficient when compared to nanoparticle transporters. This review focuses on CRISPR/Cas RNP packaging and delivery using retro/lentiviral particles and exosomes. First, we briefly describe the natural stages of viral and exosomal particle formation, release and entry into the target cells. This helps us understand the mechanisms of CRISPR/Cas RNP packaging and uncoating utilized by the current delivery systems, which we discuss afterward. Much attention is given to the exosomes released during viral particle production that can be passively loaded with RNPs as well as the mechanisms necessary for particle fusion, RNP release, and transportation inside the target cells. Collectively, together with specific packaging mechanisms, all these factors can substantially influence the editing efficiency of the system. Finally, we discuss ways to improve CRISPR/Cas RNP delivery using extracellular nanoparticles.
Collapse
Affiliation(s)
- Dmitriy Mazurov
- Cell and Gene Technology Group, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology RAS, 119334 Moscow, Russia
- Correspondence: or
| | - Lama Ramadan
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141701 Moscow, Russia
| | - Natalia Kruglova
- Cell and Gene Technology Group, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology RAS, 119334 Moscow, Russia
| |
Collapse
|
25
|
Burton JB, Carruthers NJ, Stemmer PM. Enriching extracellular vesicles for mass spectrometry. MASS SPECTROMETRY REVIEWS 2023; 42:779-795. [PMID: 34632607 DOI: 10.1002/mas.21738] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 09/19/2021] [Accepted: 09/24/2021] [Indexed: 06/13/2023]
Abstract
Extracellular vesicles from plasma, other body fluids and cell culture media hold great promise in the search for biomarkers. Exosomes in particular, the vesicle type that is secreted after being produced in the endocytic pathway and having a diameter of 30-150 nm, are considered to be a conveyance for signaling molecules and, therefore, to hold valuable information regarding the health and activity status of the cells from which they are released. The vesicular nature of exosomes is central to all methods used to separate them from the highly abundant proteins in plasma and other fluids. The enrichment of the vesicles is essential for mass spectrometry-based analysis as they represent only a very small component of all plasma proteins. The progression of isolation techniques for exosomes from ultracentrifugation through chromatographic separation using hydrophobic packing materials shows that effective enrichment is possible and that high throughput approaches to exosome enrichment are achievable.
Collapse
Affiliation(s)
- Jordan B Burton
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan, USA
| | | | - Paul M Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
26
|
Mi X, Wang SC, Winters MA, Carta G. Protein adsorption on core-shell resins for flow-through purifications: Effect of protein molecular size, shape, and salt concentration. Biotechnol Prog 2023; 39:e3300. [PMID: 36101005 DOI: 10.1002/btpr.3300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/12/2022] [Accepted: 09/07/2022] [Indexed: 11/10/2022]
Abstract
This work addresses the functional properties of the core-shell resins Capto Core 400 and 700 for a broad range of proteins spanning 66.5 to 660 kDa in molecular mass, including bovine serum albumin (BSA) in monomer and dimer form, fibronectin, thyroglobulin, and BSA conjugates with 10 and 30 kDa poly(ethylene glycol) chains. Negatively charged latex nanoparticles (NPs) with nominal diameters of 20, 40, and 100 nm are also studied as surrogates for bioparticles. Protein binding and its trends with respect to salt concentration depend on the protein size and are different for the two agarose-based multimodal resins. For the smaller proteins, the amount of protein bound over practical time scales is limited by the resin surface area and is larger for Capto Core 400 compared with Capto Core 700. For the larger proteins, diffusion is severely restricted in Capto Core 400, resulting in lower binding capacities than those observed for Capto Core 700 despite the larger surface area. Adding 500 mM NaCl reduces the local bound protein concentration and diffusional hindrance resulting in higher binding capacities for the large proteins in Capto Core 400 compared with low ionic strength conditions. The NPs are essentially completely excluded from the Capto Core 400 pores. However, 20 and 40 nm NPs bind significantly to Capto Core 700, further hindering protein diffusion. A model is provided to predict the dynamic binding capacities as a function of residence time.
Collapse
Affiliation(s)
- Xue Mi
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Sheng-Ching Wang
- Vaccine Process Research & Development, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Michael A Winters
- Vaccine Process Research & Development, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Giorgio Carta
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
27
|
Silva RM, Sousa Rosa S, Cunha R, Lobato da Silva C, Azevedo AM, Fernandes-Platzgummer A. Anion exchange chromatography-based platform for the scalable purification of extracellular vesicles derived from human mesenchymal stromal cells. Sep Purif Technol 2023. [DOI: 10.1016/j.seppur.2023.123238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
28
|
Manufactured extracellular vesicles as human therapeutics: challenges, advances, and opportunities. Curr Opin Biotechnol 2022; 77:102776. [PMID: 36041354 DOI: 10.1016/j.copbio.2022.102776] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/05/2022] [Accepted: 07/24/2022] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) have evolved across all phyla as an intercellular communication system. There are intrinsic advantages of leveraging this capability to deliver therapeutic cargo to treat disease, which have been demonstrated in numerous in vivo studies. As with other new modalities, the challenge has now shifted from proof of concept to developing reliable and efficient large-scale infrastructure to manufacture consistently pure and potent drug for broad-based patient access. This review focuses on how this challenge has been met with both existing and emerging technology platforms that are making impressive strides in the industrialization of EV manufacturing. In addition, we also highlight the gaps and opportunities that are beginning to be explored and addressed to hasten ushering in the era of therapeutic EVs.
Collapse
|
29
|
Gritti F. Modeling of the transient diffusion regime in fully porous particles - Application to the analysis of large biomolecules by ultra-high pressure liquid chromatography. J Chromatogr A 2022; 1679:463362. [DOI: 10.1016/j.chroma.2022.463362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022]
|
30
|
Barnes B, Caws T, Thomas S, Shephard AP, Corteling R, Hole P, Bracewell DG. Investigating heparin affinity chromatography for extracellular vesicle purification and fractionation. J Chromatogr A 2022; 1670:462987. [DOI: 10.1016/j.chroma.2022.462987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/02/2022] [Accepted: 03/18/2022] [Indexed: 01/15/2023]
|
31
|
A scalable, integrated downstream process for production of a recombinant measles virus-vectored vaccine. Vaccine 2022; 40:1323-1333. [PMID: 35094870 DOI: 10.1016/j.vaccine.2022.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/16/2021] [Accepted: 01/07/2022] [Indexed: 11/20/2022]
Abstract
Purification of very large and complex, enveloped viruses, such as measles virus is very challenging, it must be performed in a closed system because the final product cannot be sterile filtered and often loss of virus titer and poor product purity has been observed. We developed a purification process where the clarified and endonuclease treated culture supernatant is loaded on a restricted access chromatography medium where small impurities are bound and the virus is collected in the flow-through, which is then concentrated, and buffer exchanged by ultra/diafiltration. Up to 98.5% of host cell proteins could be captured by direct loading of clarified and endonuclease treated cell culture supernatant. Reproducible process performance and scalability of the chromatography step were demonstrated from small to pilot scale, including loading volumes from 50 mL up to 9 L. A 10-fold virus concentration was achieved by the ultrafiltration using a 100 kDa flat-sheet membrane. The order of individual process steps had a large impact on the virus infectivity and total process yields. The developed process maintained virus infectivity and is twice as fast as the traditional process train, where concentration is performed before loading on the chromatography column. Capturing impurities by the restricted access medium makes it a platform purification process with a high flexibility, which can be easily and quickly adapted to other vectors based on the measles virus vector platform.
Collapse
|
32
|
Lavado-García J, Zhang T, Cervera L, Gòdia F, Wuhrer M. Differential N- and O-glycosylation signatures of HIV-1 Gag virus-like particles and coproduced extracellular vesicles. Biotechnol Bioeng 2022; 119:1207-1221. [PMID: 35112714 PMCID: PMC9303603 DOI: 10.1002/bit.28051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 11/08/2022]
Abstract
HIV-1 virus-like particles (VLPs) are nanostructures derived from the self-assembly and cell budding of Gag polyprotein. Mimicking the native structure of the virus and being non-infectious, they represent promising candidates for the development of new vaccines as they elicit a strong immune response. In addition to this, the bounding membrane can be functionalized with exogenous antigens to target different diseases. Protein glycosylation depends strictly on the production platform and expression system used and the displayed glycosylation patterns may influence down-stream processing as well as the immune response. One of the main challenges for the development of Gag VLP production bioprocess is the separation of VLPs and coproduced extracellular vesicles (EVs). In this work, porous graphitized carbon separation method coupled with mass spectrometry was used to characterize the N- and O- glycosylation profiles of Gag VLPs produced in HEK293 cells. We identified differential glycan signatures between VLPs and EVs that could pave the way for further separation and purification strategies in order to optimize downstream processing and move forward in VLP-based vaccine production technology. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jesús Lavado-García
- Grup d'Enginyeria Cel·lular i Bioprocessos, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Laura Cervera
- Grup d'Enginyeria Cel·lular i Bioprocessos, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Francesc Gòdia
- Grup d'Enginyeria Cel·lular i Bioprocessos, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
33
|
Fuks PE, Carta G. Preparation and characterization of agarose-encapsulated ceramic hydroxyapatite particles for flow-through chromatography. SEP SCI TECHNOL 2022. [DOI: 10.1080/01496395.2022.2026388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Preston E. Fuks
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Giorgio Carta
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
34
|
Particles in Biopharmaceutical Formulations, Part 2: An Update on Analytical Techniques and Applications for Therapeutic Proteins, Viruses, Vaccines and Cells. J Pharm Sci 2021; 111:933-950. [PMID: 34919969 DOI: 10.1016/j.xphs.2021.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 11/21/2022]
Abstract
Particles in biopharmaceutical formulations remain a hot topic in drug product development. With new product classes emerging it is crucial to discriminate particulate active pharmaceutical ingredients from particulate impurities. Technical improvements, new analytical developments and emerging tools (e.g., machine learning tools) increase the amount of information generated for particles. For a proper interpretation and judgment of the generated data a thorough understanding of the measurement principle, suitable application fields and potential limitations and pitfalls is required. Our review provides a comprehensive overview of novel particle analysis techniques emerging in the last decade for particulate impurities in therapeutic protein formulations (protein-related, excipient-related and primary packaging material-related), as well as particulate biopharmaceutical formulations (virus particles, virus-like particles, lipid nanoparticles and cell-based medicinal products). In addition, we review the literature on applications, describe specific analytical approaches and illustrate advantages and drawbacks of currently available techniques for particulate biopharmaceutical formulations.
Collapse
|
35
|
González-Félix MA, Mejía-Manzano LA, González-Valdez J. Biological nanoparticles: Relevance as novel target drug delivery systems and leading chromatographic isolation approaches. Electrophoresis 2021; 43:109-118. [PMID: 34791693 DOI: 10.1002/elps.202100124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/23/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022]
Abstract
Nanotechnology is one of the most promising technologies of the 21st century, and it is now presenting an enormous impact on target drug delivery. In this context, the recent use of natural vesicle-like nanoparticles such as extracellular vesicles (i.e., exosomes, microvesicles, and apoptotic bodies) and virus-like particles is rendering encouraging results mostly because these delivery systems present cargo versatility, favorable body circulating advantages, biocompatibility, immunogenicity, and the capacity to be modified superficially to increase their affinity to a certain target or to control their entrance to the cell. However, some of the biggest challenges toward their clinical implementation are poorly standardized processing operations due to their inherent heterogeneity and expensive, long-lasting, and difficult to scale isolation procedures that can also affect the stability of the particles. Under these circumstances, chromatographic procedures represent an attractive and favorable alternative to overcome their downstream processing. Moreover, even when standardized chromatographic purification protocols are still in development, great achievements have been made using size exclusion, ionic exchange, hydrophobic interaction, and affinity protocols, mostly because of the correct harnessing of the nanovesicle membrane properties. In this sense, this review focuses on presenting the current understanding on the most promising therapeutic biological nanoparticles and the chromatographic isolation approaches employed in their recovery, providing at the same time recent findings and a general overview of the aspects that might impact the outcome of chromatographic techniques for this application.
Collapse
|
36
|
Chen C, Wang J, Sun M, Li J, Wang HMD. Toward the next-generation phyto-nanomedicines: cell-derived nanovesicles (CDNs) for natural product delivery. Biomed Pharmacother 2021; 145:112416. [PMID: 34781147 DOI: 10.1016/j.biopha.2021.112416] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 02/08/2023] Open
Abstract
Phytochemicals are plant-derived bioactive compounds, which have been widely used for therapeutic purposes. Due to the poor water-solubility, low bioavailability and non-specific targeting characteristic, diverse classes of nanocarriers are utilized for encapsulation and delivery of bio-effective agents. Cell-derived nanovesicles (CDNs), known for exosomes or extracellular vesicles (EVs), are biological nanoparticles with multiple functions. Compared to the artificial counterpart, CDNs hold great potential in drug delivery given the higher stability, superior biocompatibility and the lager capability of encapsulating bioactive molecules. Here, we provide a bench-to-bedside review of CDNs-based nanoplatform, including the bio-origin, preparation, characterization and functionalization. Beyond that, the focus is laid on the therapeutic effect of CDNs-mediated drug delivery for natural products. The state-of-art development as well as some pre-clinical applications of using CDNs for disease treatment is also summarized. It is highly expected that the continuing development of CDNs-based delivery systems will further promote the clinical utilization and translation of phyto-nanomedicines.
Collapse
Affiliation(s)
- Chaoxiang Chen
- College of Food and Biological Engineering, Jimei University, China
| | - Jialin Wang
- College of Food and Biological Engineering, Jimei University, China
| | - Mengdi Sun
- College of Food and Biological Engineering, Jimei University, China
| | - Jian Li
- College of Food and Biological Engineering, Jimei University, China.
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
37
|
Dosing extracellular vesicles. Adv Drug Deliv Rev 2021; 178:113961. [PMID: 34481030 DOI: 10.1016/j.addr.2021.113961] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/19/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are natural nanoparticles containing biologically active molecules. They are important mediators of intercellular communication and can be exploited therapeutically by various bioengineering approaches. To accurately determine the therapeutic potential of EVs in pre-clinical and clinical settings, dependable dosing strategies are of utmost importance. However, the field suffers from inconsistencies comprising all areas of EV production and characterisation. Therefore, a standardised and well-defined process in EV quantification, key to reliable therapeutic EV dosing, remains to be established. Here, we examined 64 pre-clinical studies for EV-based therapeutics with respect to their applied EV dosing strategies. We identified variations in effective dosing strategies irrespective of the applied EV purification method and cell source. Moreover, we found dose discrepancies depending on the disease model, where EV doses were selected without accounting for published EV pharmacokinetics or biodistribution patterns. We therefore propose to focus on qualitative aspects when dosing EV-based therapeutics, such as the potency of the therapeutic cargo entity. This will ensure batch-to-batch reliability and enhance reproducibility between applications. Furthermore, it will allow for the successful benchmarking of EV-based therapeutics compared to other nanoparticle drug delivery systems, such as viral vector-based or lipid-based nanoparticle approaches.
Collapse
|
38
|
González-Domínguez I, Lorenzo E, Bernier A, Cervera L, Gòdia F, Kamen A. A Four-Step Purification Process for Gag VLPs: From Culture Supernatant to High-Purity Lyophilized Particles. Vaccines (Basel) 2021; 9:vaccines9101154. [PMID: 34696262 PMCID: PMC8539588 DOI: 10.3390/vaccines9101154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 01/15/2023] Open
Abstract
Gag-based virus-like particles (VLPs) have high potential as scaffolds for the development of chimeric vaccines and delivery strategies. The production of purified preparations that can be preserved independently from cold chains is highly desirable to facilitate distribution and access worldwide. In this work, a nimble purification has been developed, facilitating the production of Gag VLPs. Suspension-adapted HEK 293 cells cultured in chemically defined cell culture media were used to produce the VLPs. A four-step downstream process (DSP) consisting of membrane filtration, ion-exchange chromatography, polishing, and lyophilization was developed. The purification of VLPs from other contaminants such as host cell proteins (HCP), double-stranded DNA, or extracellular vesicles (EVs) was confirmed after their DSP. A concentration of 2.2 ± 0.8 × 109 VLPs/mL in the lyophilized samples was obtained after its storage at room temperature for two months. Morphology and structural integrity of purified VLPs was assessed by cryo-TEM and NTA. Likewise, the purification methodologies proposed here could be easily scaled up and applied to purify similar enveloped viruses and vesicles.
Collapse
Affiliation(s)
- Irene González-Domínguez
- Departament d’Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.L.); (L.C.); (F.G.)
- Correspondence:
| | - Elianet Lorenzo
- Departament d’Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.L.); (L.C.); (F.G.)
| | - Alice Bernier
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada; (A.B.); (A.K.)
| | - Laura Cervera
- Departament d’Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.L.); (L.C.); (F.G.)
| | - Francesc Gòdia
- Departament d’Enginyeria Química Biològica i Ambiental, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (E.L.); (L.C.); (F.G.)
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada; (A.B.); (A.K.)
| |
Collapse
|
39
|
Li G, Ma W, Mo J, Cheng B, Shoda SI, Zhou D, Ye XS. Influenza Virus Precision Diagnosis and Continuous Purification Enabled by Neuraminidase-Resistant Glycopolymer-Coated Microbeads. ACS APPLIED MATERIALS & INTERFACES 2021; 13:46260-46269. [PMID: 34547894 DOI: 10.1021/acsami.1c11561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Rapid diagnosis and vaccine development are critical to prevent the threat posed by viruses. However, rapid tests, such as colloidal gold assays, yield false-negative results due to the low quantities of viruses; moreover, conventional virus purification, including ultracentrifugation and nanofiltration, is multistep and time-consuming, which limits laboratory research and commercial development of viral vaccines. A rapid virus enrichment and purification technique will improve clinical diagnosis sensitivity and simplify vaccine production. Hence, we developed the surface-glycosylated microbeads (glycobeads) featuring chemically synthetic glycoclusters and reversible linkers to selectively capture the influenza virus. The surface plasmon resonance (SPR) evaluation indicated broad spectrum affinity of S-linked glycosides to various influenza viruses. The magnetic glycobeads were integrated into clinical rapid diagnosis, leading to a 30-fold lower limit of detection. Additionally, the captured viruses can be released under physiological conditions, delivering purified viruses with >50% recovery and without decreasing their native infectivity. Notably, this glycobead platform will facilitate the sensitive detection and continuous one-step purification of the target virus that contributes to future vaccine production.
Collapse
Affiliation(s)
- Gefei Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Wenxiao Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Juan Mo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Boyang Cheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Shin-Ichiro Shoda
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, 6-6-11, Aoba, Aoba-ku, Sendai 980-8579, Japan
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Xue Yuan Road No. 38, Beijing 100191, China
| |
Collapse
|
40
|
Scaled preparation of extracellular vesicles from conditioned media. Adv Drug Deliv Rev 2021; 177:113940. [PMID: 34419502 DOI: 10.1016/j.addr.2021.113940] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/13/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) especially of mesenchymal stem/stomal cells (MSCs) are increasingly considered as biotherapeutic agents for a variety of different diseases. For translating them effectively into the clinics, scalable production processes fulfilling good manufacturing practice (GMP) are needed. Like for other biotherapeutic agents, the manufacturing of EV products can be subdivided in the upstream and downstream processing and the subsequent quality control, each of them containing several unit operations. During upstream processing (USP), cells are isolated, stored (cell banking) and expanded; furthermore, EV-containing conditioned media are produced. During downstream processing (DSP), conditioned media (CM) are processed to obtain concentrated and purified EV products. CM are either stored until DSP or are directly processed. As first unit operation in DSP, clarification removes remaining cells, debris and other larger impurities. The key operations of each EV DSP is volume-reduction combined with purification of the concentrated EVs. Most of the EV preparation methods used in conventional research labs including differential centrifugation procedures are limited in their scalability. Consequently, it is a major challenge in the therapeutic EV field to identify appropriate EV concentration and purification methods allowing scale up. As EVs share several features with enveloped viruses, that are used for more than two decades in the clinics now, several principles can be adopted to EV manufacturing. Here, we introduce and discuss volume reducing and purification methods frequently used for viruses and analyze their value for the manufacturing of EV-based therapeutics.
Collapse
|
41
|
Buschmann D, Mussack V, Byrd JB. Separation, characterization, and standardization of extracellular vesicles for drug delivery applications. Adv Drug Deliv Rev 2021; 174:348-368. [PMID: 33964356 PMCID: PMC8217305 DOI: 10.1016/j.addr.2021.04.027] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are membranous nanovesicles secreted from living cells, shuttling macromolecules in intercellular communication and potentially possessing intrinsic therapeutic activity. Due to their stability, low immunogenicity, and inherent interaction with recipient cells, EVs also hold great promise as drug delivery vehicles. Indeed, they have been used to deliver nucleic acids, proteins, and small molecules in preclinical investigations. Furthermore, EV-based drugs have entered early clinical trials for cancer or neurodegenerative diseases. Despite their appeal as delivery vectors, however, EV-based drug delivery progress has been hampered by heterogeneity of sample types and methods as well as a persistent lack of standardization, validation, and comprehensive reporting. This review highlights specific requirements for EVs in drug delivery and describes the most pertinent approaches for separation and characterization. Despite residual uncertainties related to pharmacodynamics, pharmacokinetics, and potential off-target effects, clinical-grade, high-potency EV drugs might be achievable through GMP-compliant workflows in a highly standardized environment.
Collapse
Affiliation(s)
- Dominik Buschmann
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Veronika Mussack
- Department of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - James Brian Byrd
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
42
|
Mi X, Fuks P, Wang SC, Winters MA, Carta G. Protein Adsorption on Core-shell Particles: Comparison of Capto™ Core 400 and 700 Resins. J Chromatogr A 2021; 1651:462314. [PMID: 34144396 DOI: 10.1016/j.chroma.2021.462314] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 01/15/2023]
Abstract
Structural and functional characteristics of the two core-shell resins Capto™ Core 400 and 700, which are useful for the flow-through purification of bioparticles such as viruses, viral vectors, and vaccines, are compared using bovine serum albumin (BSA) and thyroglobulin (Tg) as models for small and large protein contaminants. Both resins are agarose-based and contain an adsorbing core surrounded by an inert shell. Although shell thicknesses are comparable (3.6 and 4.2 µm for Capto Core 400 and 700, respectively), the two resins differ substantially in pore size (pore radii of 19 and 50 nm, respectively). Because of the smaller pores and higher surface area, the BSA binding capacity of Capto Core 400 is approximately double that of Capto Core 700. However, for the much larger Tg, the attainable capacity is substantially larger for Capto Core 700. Mass transfer in both resins is affected by diffusional resistances through the shell and within the adsorbing core. For BSA, core and shell effective pore diffusivities are about 0.25 × 10-7 and 0.6 × 10-7 cm2/s, respectively, for Capto Core 400, and about 1.6 × 10-7 and 2.6 × 10-7 cm2/s, respectively, for Capto Core 700. These values decrease dramatically for Tg to 0.022 × 10-7 and 0.088 × 10-7 cm2/s and to 0.13 × 10-7 and 0.59 × 10-7 cm2/s for Capto Core 400 and 700, respectively. Adsorbed Tg further hinders diffusion of BSA in both resins. Column measurements show that, despite the higher static capacity of Capto Core 400 for BSA, the dynamic binding capacity is greater for Capto Core 700 as a result of its faster kinetics. However, some of this advantage is lost if the feed is a mixture of BSA and Tg since, in this case, Tg binding leads to greater diffusional hindrance for BSA.
Collapse
Affiliation(s)
- Xue Mi
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Preston Fuks
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Sheng-Ching Wang
- Vaccine Process Research and Development, Merck & Co., Inc., West Point, PA, USA
| | - Michael A Winters
- Vaccine Process Research and Development, Merck & Co., Inc., West Point, PA, USA
| | - Giorgio Carta
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
43
|
Xue F, Chen Y, Wen Y, Abhange K, Zhang W, Cheng G, Quinn Z, Mao W, Wan Y. Isolation of extracellular vesicles with multivalent aptamers. Analyst 2021; 146:253-261. [PMID: 33107503 DOI: 10.1039/d0an01420f] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are lipid-enclosed submicron-sized vesicles that are secreted by all eukaryotic cells. EVs can selectively encapsulate tissue-specific small molecules from parent cells and efficiently deliver them to recipient cells. As signal mediators of intercellular communication, the molecules packaged in EVs play critical roles in the pathophysiology of diseases. In relevant clinical translation, EV contents have been used for cancer diagnosis and treatment monitoring. To further promote EV-based cancer liquid biopsy toward large-scale clinical implementation, the efficient and specific isolation of pure tumor-derived EVs from body fluids is a prerequisite. However, the existing EV isolation methods are unable to address certain technical challenges, such as lengthy procedures, low throughput, low specificity, heavy protein contamination, etc., and thus, new approaches for EV isolation are required. Here, we report a multivalent, long single-stranded aptamer with repeated units for EV enrichment and retrieval. After short incubation of biotin-labeled multivalent aptamers (MAs) with the samples, EVs can be quickly secured by MAs, anchored onto streptavidin-coated microspheres, and further retrieved via digestion of the DNA aptamer. Approximately 45% of EVs can be isolated from the spiked samples in 40 min with a depletion of 84.7% of albumin contamination. In addition, 93.1% of the isolated EVs can be retrieved via DNase-mediated aptamer degradation in 10 min for downstream molecular analyses. Our findings suggest that MAs can efficiently and specifically isolate EVs derived from malignant lymphocytes, and this simple method could facilitate the EV-centered study of acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Fei Xue
- The Pq Laboratory of Micro/Nano BiomeDx, Department of Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY 13902, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vaillancourt M, Hubert A, Subra C, Boucher J, Bazié WW, Vitry J, Berrazouane S, Routy JP, Trottier S, Tremblay C, Jenabian MA, Benmoussa A, Provost P, Tessier PA, Gilbert C. Velocity Gradient Separation Reveals a New Extracellular Vesicle Population Enriched in miR-155 and Mitochondrial DNA. Pathogens 2021; 10:526. [PMID: 33925397 PMCID: PMC8146806 DOI: 10.3390/pathogens10050526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/10/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) and their contents (proteins, lipids, messenger RNA, microRNA, and DNA) are viewed as intercellular signals, cell-transforming agents, and shelters for viruses that allow both diagnostic and therapeutic interventions. EVs circulating in the blood of individuals infected with human immunodeficiency virus (HIV-1) may provide insights into pathogenesis, inflammation, and disease progression. However, distinguishing plasma membrane EVs from exosomes, exomeres, apoptotic bodies, virions, and contaminating proteins remains challenging. We aimed at comparing sucrose and iodixanol density and velocity gradients along with commercial kits as a means of separating EVs from HIV particles and contaminating protein like calprotectin; and thereby evaluating the suitability of current plasma EVs analysis techniques for identifying new biomarkers of HIV-1 immune activation. Multiple analysis have been performed on HIV-1 infected cell lines, plasma from HIV-1 patients, or plasma from HIV-negative individuals spiked with HIV-1. Commercial kits, the differential centrifugation and density or velocity gradients to precipitate and separate HIV, EVs, and proteins such as calprotectin, have been used. EVs, virions, and contaminating proteins were characterized using Western blot, ELISA, RT-PCR, hydrodynamic size measurement, and enzymatic assay. Conversely to iodixanol density or velocity gradient, protein and virions co-sedimented in the same fractions of the sucrose density gradient than AChE-positive EVs. Iodixanol velocity gradient provided the optimal separation of EVs from viruses and free proteins in culture supernatants and plasma samples from a person living with HIV (PLWH) or a control and revealed a new population of large EVs enriched in microRNA miR-155 and mitochondrial DNA. Although EVs and their contents provide helpful information about several key events in HIV-1 pathogenesis, their purification and extensive characterization by velocity gradient must be investigated thoroughly before further use as biomarkers. By revealing a new population of EVs enriched in miR-155 and mitochondrial DNA, this study paves a way to increase our understanding of HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Myriam Vaillancourt
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
| | - Audrey Hubert
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
| | - Caroline Subra
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Julien Boucher
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
| | - Wilfried Wenceslas Bazié
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
- Programme de Recherche sur les Maladies Infectieuses, Centre Muraz, Institut National de Santé Publique, Bobo-Dioulasso 01 BP 390, Burkina Faso
| | - Julien Vitry
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
| | - Sofiane Berrazouane
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
| | - Jean-Pierre Routy
- Chronic Viral Illness Service and Division of Hematology, McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Sylvie Trottier
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
- Centre de Recherche du CHU de Québec, Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Cécile Tremblay
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal, Montréal, QC H3C 3J7, Canada;
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Mohammad-Ali Jenabian
- Département des Sciences Biologiques et Centre de Recherche CERMO-FC, Université du Québec à Montréal (UQAM), Montréal, QC H2L 2C4, Canada;
| | - Abderrahim Benmoussa
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
- Department of Nutrition, CHU Sainte-Justine—Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Patrick Provost
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
- Centre de Recherche du CHU de Québec, Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Philippe A. Tessier
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
- Centre de Recherche du CHU de Québec, Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Caroline Gilbert
- Centre de Recherche du CHU de Québec-Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada; (M.V.); (A.H.); (C.S.); (J.B.); (W.W.B.); (J.V.); (S.B.); (S.T.); (A.B.); (P.P.); (P.A.T.)
- Centre de Recherche du CHU de Québec, Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université Laval, T1-49, 2705 boulevard Laurier, Québec, QC G1V 4G2, Canada
| |
Collapse
|
45
|
Kip C, Hamaloğlu KÖ, Demir C, Tuncel A. Recent trends in sorbents for bioaffinity chromatography. J Sep Sci 2021; 44:1273-1291. [PMID: 33370505 DOI: 10.1002/jssc.202001117] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/24/2022]
Abstract
Isolation or enrichment of biological molecules from complex biological samples is mostly a prerequisite in proteomics, genomics, and glycomics. Different techniques have been used to advance the efficiency of the purification of biological molecules. Bioaffinity chromatography is one of the most powerful technique that plays an important role in the isolation of target biological molecules by the specific interactions with ligands that are immobilized on different support materials. This review examines the recent developments in bioaffinity chromatography particularly over the past 5 years in the literature. Also properties of supports, immobilization techniques, types of binding agents, and methods used in bioaffinity chromatography applications are summarized.
Collapse
Affiliation(s)
- Cigdem Kip
- Chemical Engineering Department, Hacettepe University, Ankara, Turkey
| | | | - Cihan Demir
- Chemical Engineering Department, Hacettepe University, Ankara, Turkey.,Nanotechnology and Nanomedicine Division, Hacettepe University, Ankara, Turkey
| | - Ali Tuncel
- Chemical Engineering Department, Hacettepe University, Ankara, Turkey
| |
Collapse
|
46
|
Puente-Massaguer E, Grau-Garcia P, Strobl F, Grabherr R, Striedner G, Lecina M, Gòdia F. Accelerating HIV-1 VLP production using stable High Five insect cell pools. Biotechnol J 2020; 16:e2000391. [PMID: 33247883 DOI: 10.1002/biot.202000391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/21/2020] [Indexed: 12/25/2022]
Abstract
Stable cell pools are receiving a renewed interest as a potential alternative system to clonal cell lines. The shorter development timelines and the capacity to achieve high product yields make them an interesting approach for recombinant protein production. In this study, stable High Five cell pools are assessed for the production of a simple protein, mCherry, and the more complex HIV-1 Gag-eGFP virus-like particles (VLPs). Random integration coupled to fluorescence-activated cell sorting (FACS) in suspension conditions is applied to accelerate the stable cell pool generation process and enrich it with high producer cells. This methodology is successfully transferred to a bioreactor for VLP production, resulting in a 2-fold increase in VLP yields with respect to shake flask cultures. In these conditions, maximum viable cell concentration improves by 1.5-fold, and by-product formation is significantly reduced. Remarkably, a global increase in the uptake of amino acids in the Gag-eGFP stable cell pool is observed when compared with parental High Five cells, reflecting the additional metabolic burden associated with VLP production. These results suggest that stable High Five cell pools are a robust and powerful approach to produce VLPs and other recombinant proteins, and put the basis for future studies aiming to scale up this system.
Collapse
Affiliation(s)
- Eduard Puente-Massaguer
- Departament d'Enginyeria Química, Biològica i Ambiental, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, 08193, Spain
| | - Paula Grau-Garcia
- Departament d'Enginyeria Química, Biològica i Ambiental, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, 08193, Spain
| | - Florian Strobl
- Austrian Centre of Industrial Biotechnology (acib GmbH), Vienna, 1010, Austria.,Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Reingard Grabherr
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Gerald Striedner
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Martí Lecina
- IQS School of Engineering, Universitat Ramón Llull, Barcelona, 08017, Spain
| | - Francesc Gòdia
- Departament d'Enginyeria Química, Biològica i Ambiental, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, 08193, Spain
| |
Collapse
|
47
|
Jung S, Jacobs KFK, Shein M, Schütz AK, Mohr F, Stadler H, Stadler D, Lucko AM, Altstetter SM, Wilsch F, Deng L, Protzer U. Efficient and reproducible depletion of hepatitis B virus from plasma derived extracellular vesicles. J Extracell Vesicles 2020; 10:e12040. [PMID: 33363711 PMCID: PMC7754750 DOI: 10.1002/jev2.12040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 09/24/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging fundamental players in viral infections by shuttling viral components, mediating immune responses and likely the spread of the virus. However, the obstacles involved in purifying EVs and removing contaminating viral particles in a reliable and effective manner bottlenecks the full potential for the development of clinical and diagnostic treatment options targeting EV. Because of the similarities in size, density, membrane composition and mode of biogenesis of EVs and virions there are no standardized approaches for virus-removal from EV preparations yet. Functional EV studies also require EV samples that are devoid of antibody contaminants. Consequently, the study of EVs in virology needs reliable and effective protocols to purify EVs and remove contaminating antibodies and viral particles. Here, we established a protocol for EV purification from hepatitis B virus (HBV)-containing plasma by a combination of size-exclusion chromatography and affinity-based purification. After purification, EV samples were free of virus-sized particles, HBV surface antigen, HBV core antigen, antibodies or infectious material. Viral genomic contamination was also decreased following purification. By using appropriate antibodies and size parameters, this protocol could potentially be applied to purification of EVs from other viral samples. In summary, we established a fast, reproducible and robust approach for the removal of HBV from EV preparations. Looking forward to the point of purifying EVs from clinical samples, this method should enable studies shedding light on the underlying mechanisms of EVs in viral infections and their diagnostic and prognostic potential.
Collapse
Affiliation(s)
- Stephanie Jung
- Institute of VirologySchool of MedicineHelmholtz Zentrum München/Technical University of MunichGarchingGermany
| | | | - Mikhail Shein
- Bavarian NMR Center, Department of ChemistryTechnical University of MunichGarchingGermany
| | - Anne Kathrin Schütz
- Bavarian NMR Center, Department of ChemistryTechnical University of MunichGarchingGermany
| | | | | | - Daniela Stadler
- Institute of VirologySchool of MedicineHelmholtz Zentrum München/Technical University of MunichGarchingGermany
| | - Aaron Michael Lucko
- Institute of VirologySchool of MedicineHelmholtz Zentrum München/Technical University of MunichGarchingGermany
| | | | - Florian Wilsch
- Institute of VirologySchool of MedicineHelmholtz Zentrum München/Technical University of MunichGarchingGermany
| | - Li Deng
- Institute of VirologySchool of MedicineHelmholtz Zentrum München/Technical University of MunichGarchingGermany
| | - Ulrike Protzer
- Institute of VirologySchool of MedicineHelmholtz Zentrum München/Technical University of MunichGarchingGermany
- German Center for Infection Research (DZIF)Munich partner siteGarchingGermany
| |
Collapse
|
48
|
Lorenc T, Chrzanowski J, Olejarz W. Current Perspectives on Clinical Use of Exosomes as a Personalized Contrast Media and Theranostics. Cancers (Basel) 2020; 12:E3386. [PMID: 33207614 PMCID: PMC7698051 DOI: 10.3390/cancers12113386] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/08/2020] [Accepted: 11/14/2020] [Indexed: 02/07/2023] Open
Abstract
An appropriate combination of biomarkers and imaging technologies will become standard practice in the future. Because the incidence of and mortality from cancers is rising, the further study of new approaches for the early detection and precise characterization of tumors is essential. Extracellular vesicles (EVs), including exosomes, prove to have great potential when it comes to diagnosis and targeted therapy. Due to their natural ability to pass through biological barriers, depending on their origin, EVs can accumulate at defined sites, including tumors, preferentially. This manuscript discusses the difficulties and simplicities of processing cell-derived materials, packaging diverse groups of agents in EVs, and activating the biological complex. Developing exosome-based diagnostic techniques to detect disease precisely and early as well as treat disease marks a new era of personalized radiology and nuclear medicine. As circulating drug delivery vehicles for novel therapeutic modalities, EVs offer a new platform for cancer theranostic.
Collapse
Affiliation(s)
- Tomasz Lorenc
- Ist Department of Clinical Radiology, Medical University of Warsaw, 5 Chalubinskiego Street, 02-004 Warsaw, Poland
| | - Julian Chrzanowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (J.C.); (W.O.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland; (J.C.); (W.O.)
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
49
|
Lavado-García J, González-Domínguez I, Cervera L, Jorge I, Vázquez J, Gòdia F. Molecular Characterization of the Coproduced Extracellular Vesicles in HEK293 during Virus-Like Particle Production. J Proteome Res 2020; 19:4516-4532. [PMID: 32975947 PMCID: PMC7640977 DOI: 10.1021/acs.jproteome.0c00581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Indexed: 12/22/2022]
Abstract
Vaccine therapies based on virus-like particles (VLPs) are currently in the spotlight due to their potential for generating high immunogenic responses while presenting fewer side effects than conventional vaccines. These self-assembled nanostructures resemble the native conformation of the virus but lack genetic material. They are becoming a promising platform for vaccine candidates against several diseases due to the ability of modifying their membrane with antigens from different viruses. The coproduction of extracellular vesicles (EVs) when producing VLPs is a key phenomenon currently still under study. In order to characterize this extracellular environment, a quantitative proteomics approach has been carried out. Three conditions were studied: non-transfected, transfected with an empty plasmid as control, and transfected with a plasmid coding for HIV-1 Gag polyprotein. A shift in EV biogenesis has been detected upon transfection, changing the production from large to small EVs. Another remarkable trait found was the presence of DNA being secreted within vesicles smaller than 200 nm. Studying the protein profile of these biological nanocarriers, it was observed that EVs were reflecting an overall energy homeostasis disruption via mitochondrial protein deregulation. Also, immunomodulatory proteins like ITGB1, ENO3, and PRDX5 were identified and quantified in VLP and EV fractions. These findings provide insight on the nature of the VLP extracellular environment defining the characteristics and protein profile of EVs, with potential to develop new downstream separation strategies or using them as adjuvants in viral therapies.
Collapse
Affiliation(s)
- Jesús Lavado-García
- Grup
d’Enginyeria Cellular i Bioprocés, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola
del Vallès, 08193 Barcelona, Spain
| | - Irene González-Domínguez
- Grup
d’Enginyeria Cellular i Bioprocés, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola
del Vallès, 08193 Barcelona, Spain
| | - Laura Cervera
- Grup
d’Enginyeria Cellular i Bioprocés, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola
del Vallès, 08193 Barcelona, Spain
| | - Inmaculada Jorge
- Laboratory
of Cardiovascular Proteomics, Centro Nacional
Investigaciones Cardiovasculares (CNIC), C/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Centro
de Investigación Biomédica en Red Enfermedades Cardiovasculares
(CIBERCV), Madrid, Spain
| | - Jesús Vázquez
- Laboratory
of Cardiovascular Proteomics, Centro Nacional
Investigaciones Cardiovasculares (CNIC), C/Melchor Fernández Almagro 3, Madrid 28029, Spain
- Centro
de Investigación Biomédica en Red Enfermedades Cardiovasculares
(CIBERCV), Madrid, Spain
| | - Francesc Gòdia
- Grup
d’Enginyeria Cellular i Bioprocés, Escola d’Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola
del Vallès, 08193 Barcelona, Spain
| |
Collapse
|
50
|
Martínez-Molina E, Chocarro-Wrona C, Martínez-Moreno D, Marchal JA, Boulaiz H. Large-Scale Production of Lentiviral Vectors: Current Perspectives and Challenges. Pharmaceutics 2020; 12:pharmaceutics12111051. [PMID: 33153183 PMCID: PMC7693937 DOI: 10.3390/pharmaceutics12111051] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/20/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023] Open
Abstract
Lentiviral vectors (LVs) have gained value over recent years as gene carriers in gene therapy. These viral vectors are safer than what was previously being used for gene transfer and are capable of infecting both dividing and nondividing cells with a long-term expression. This characteristic makes LVs ideal for clinical research, as has been demonstrated with the approval of lentivirus-based gene therapies from the Food and Drug Administration and the European Agency for Medicine. A large number of functional lentiviral particles are required for clinical trials, and large-scale production has been challenging. Therefore, efforts are focused on solving the drawbacks associated with the production and purification of LVsunder current good manufacturing practice. In recent years, we have witnessed the development and optimization of new protocols, packaging cell lines, and culture devices that are very close to reaching the target production level. Here, we review the most recent, efficient, and promising methods for the clinical-scale production ofLVs.
Collapse
Affiliation(s)
- Eduardo Martínez-Molina
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
| | - Carlos Chocarro-Wrona
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain
| | - Daniel Martínez-Moreno
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain
| | - Juan A. Marchal
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain
| | - Houria Boulaiz
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada (D.M.), 18016 Granada, Spain; (E.M.-M.); (C.C.-W.); (D.M.-M.); (J.A.M.)
- Department of Human Anatomy and Embryology, University of Granada, 18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, 18016 Granada, Spain
- Correspondence: ; Tel.: +34-958-241-271
| |
Collapse
|