1
|
Sobczak JM, Barkovska I, Balke I, Rothen DA, Mohsen MO, Skrastina D, Ogrina A, Martina B, Jansons J, Bogans J, Vogel M, Bachmann MF, Zeltins A. Identifying Key Drivers of Efficient B Cell Responses: On the Role of T Help, Antigen-Organization, and Toll-like Receptor Stimulation for Generating a Neutralizing Anti-Dengue Virus Response. Vaccines (Basel) 2024; 12:661. [PMID: 38932390 PMCID: PMC11209419 DOI: 10.3390/vaccines12060661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
T help (Th), stimulation of toll-like receptors (pathogen-associated molecular patterns, PAMPs), and antigen organization and repetitiveness (pathogen-associated structural patterns, PASPs) were shown numerous times to be important in driving B-cell and antibody responses. In this study, we dissected the individual contributions of these parameters using newly developed "Immune-tag" technology. As model antigens, we used eGFP and the third domain of the dengue virus 1 envelope protein (DV1 EDIII), the major target of virus-neutralizing antibodies. The respective proteins were expressed alone or genetically fused to the N-terminal fragment of the cucumber mosaic virus (CMV) capsid protein-nCMV, rendering the antigens oligomeric. In a step-by-step manner, RNA was attached as a PAMP, and/or a universal Th-cell epitope was genetically added for additional Th. Finally, a PASP was added to the constructs by displaying the antigens highly organized and repetitively on the surface of CMV-derived virus-like particles (CuMV VLPs). Sera from immunized mice demonstrated that each component contributed stepwise to the immunogenicity of both proteins. All components combined in the CuMV VLP platform induced by far the highest antibody responses. In addition, the DV1 EDIII induced high levels of DENV-1-neutralizing antibodies only if displayed on VLPs. Thus, combining multiple cues typically associated with viruses results in optimal antibody responses.
Collapse
Affiliation(s)
- Jan M. Sobczak
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Irena Barkovska
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Ina Balke
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Dominik A. Rothen
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Mona O. Mohsen
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Dace Skrastina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Anete Ogrina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Byron Martina
- Artemis Bioservices, 2629 JD Delft, The Netherlands;
- Protinhi Therapeutics, 6534 AT Nijmegen, The Netherlands
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Janis Bogans
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Monique Vogel
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| |
Collapse
|
2
|
Ughade S, Rana S, Nadeem M, Kumthekar R, Mahajani S, Bhambure R. Mechanistic Modeling of Size Exclusion Chromatography-Assisted In Vitro Refolding of the Recombinant Biosimilar Teriparatide (PTH-34). ACS OMEGA 2024; 9:3204-3216. [PMID: 38284095 PMCID: PMC10809233 DOI: 10.1021/acsomega.3c04463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/10/2023] [Accepted: 12/21/2023] [Indexed: 01/30/2024]
Abstract
In vitro protein refolding is one of the critical unit operations in manufacturing recombinant peptides expressed using Escherichia coli as host cells. This study is focused on designing size exclusion chromatography-assisted in vitro refolding process for biosimilar recombinant parathyroid hormone. Inclusion bodies (IBs) of recombinant parathyroid hormone were solubilized at higher pH, and in vitro refolding was performed using size exclusion chromatography. In the first part of the investigation, DoE-based empirical optimization was performed to achieve a higher refolding yield for a biosimilar recombinant parathyroid hormone. The effect of solubilized inclusion body (IB) feed volume, concentration of IBs, and residence time on in vitro refolding of recombinant teriparatide was studied using the Box-Behnken design. Size exclusion chromatography (SEC)-assisted in vitro refolding was performed at 8 °C at pH 10.5 by using 20 mM Tris buffer. The maximum refolding yield of 98.12% was achieved at feed volume (12.5% of CV) and 20 mg/mL inclusion body (IB) concentration with a residence time of 50 min and a purity of 66.1% based on densitometric analysis using SDS-PAGE. In the latter part of the investigation, the general rate mechanistic model framework for size exclusion chromatography was developed and validated with the experimental results. The developed model helped in the accurate prediction of the elution volumes and product yield. The developed model also helps to predict the elution performance of a scalable column a priori. Post in vitro refolding, the formation of the native peptide structure was examined using various orthogonal analytical tools to study the protein's primary, secondary, and tertiary structures. The developed hybrid process development approach is a valuable tool toachieve high-yield, scalable refolding conditions for recombinant proteins without disulfide bonds.
Collapse
Affiliation(s)
- Santosh Ughade
- Chemical Engineering and Process Development Division, CSIR - National Chemical Laboratory, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sunil Rana
- Chemical Engineering and Process Development Division, CSIR - National Chemical Laboratory, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mohd Nadeem
- Chemical Engineering and Process Development Division, CSIR - National Chemical Laboratory, Pune 411008, India
- Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Rupali Kumthekar
- Chemical Engineering and Process Development Division, CSIR - National Chemical Laboratory, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sanjay Mahajani
- Chemical Engineering and Process Development Division, CSIR - National Chemical Laboratory, Pune 411008, India
- Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Rahul Bhambure
- Chemical Engineering and Process Development Division, CSIR - National Chemical Laboratory, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
3
|
Rana S, Ughade S, Kumthekar R, Bhambure R. Chromatography assisted in-vitro refolding and purification of recombinant peptibody: Recombinant Romiplostim a case study. Int J Biol Macromol 2023; 249:126037. [PMID: 37516226 DOI: 10.1016/j.ijbiomac.2023.126037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/16/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
In-vitro protein refolding is one of the key rate-limiting unit operations in manufacturing of fusion proteins such as peptibodies expressed using E. coli. Dilution-assisted refolding is the most commonly used industrial practice to achieve the soluble, native functional form of the recombinant protein from the inclusion bodies. This study is focused on developing a chromatography-assisted in-vitro refolding platform to produce the biologically active, native form of recombinant peptibody. Recombinant Romiplostim was selected as a model protein for the study. A plug flow tubular reactor was connected in series with capture step affinity chromatography to achieve simultaneous in-vitro refolding and capture step purification of recombinant Romiplostim. Effect of various critical process parameters like fold dilution, temperature, residence time, and Cysteine: DTT ratio was studied using a central composite based design of experiment strategy to achieve a maximum refolding yield of selected peptibody. Under optimum refolding conditions, the maximum refolding yield of 57.0 ± 1.5 % and a purity of over 79.73 ± 3.4 % were achieved at 25-fold dilution, 15 °C temperature, 6 h residence time with 6 mM and 10 mM of cysteine and DTT, respectively. The formation of native peptibody structure was examined using various orthogonal analytical tools to study the protein's primary, secondary, and tertiary structure. The amino acid sequence for the disulfide-linked peptide was mapped using collision-induced dissociation (CID) to confirm the formation of interchain disulfide bonds between Cys7-Cys7 and Cys10-Cys10 similarly for intra-chain disulfide bonds between Cys42-Cys102, and Cys148-Cys206. The developed protocol here is a valuable tool to identify high-yield scalable refolding conditions for multi-domain proteins involving inter-domain disulfide bonds.
Collapse
Affiliation(s)
- Sunil Rana
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh Ughade
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rupali Kumthekar
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rahul Bhambure
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
4
|
Wu T, Song Z, Huang H, Jakos T, Jiang H, Xie Y, Zhu J. Construction and evaluation of GPC3-targeted immunotoxins as a novel therapeutic modality for hepatocellular carcinoma. Int Immunopharmacol 2022; 113:109393. [DOI: 10.1016/j.intimp.2022.109393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/11/2022] [Accepted: 10/24/2022] [Indexed: 11/15/2022]
|
5
|
Buscajoni L, Martinetz MC, Berkemeyer M, Brocard C. Refolding in the modern biopharmaceutical industry. Biotechnol Adv 2022; 61:108050. [PMID: 36252795 DOI: 10.1016/j.biotechadv.2022.108050] [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: 06/07/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/02/2022]
Abstract
Inclusion bodies (IBs) often emerge upon overexpression of recombinant proteins in E. coli. From IBs, refolding is necessary to generate the native protein that can be further purified to obtain pure and active biologicals. This work focusses on refolding as a significant process step during biopharmaceutical manufacturing with an industrial perspective. A theoretical and historical background on protein refolding gives the reader a starting point for further insights into industrial process development. Quality requirements on IBs as starting material for refolding are discussed and further economic and ecological aspects are considered with regards to buffer systems and refolding conditions. A process development roadmap shows the development of a refolding process starting from first exploratory screening rounds to scale-up and implementation in manufacturing plant. Different aspects, with a direct influence on yield, such as the selection of chemicals including pH, ionic strength, additives, etc., and other often neglected aspects, important during scale-up, such as mixing, and gas-fluid interaction, are highlighted with the use of a quality by design (QbD) approach. The benefits of simulation sciences (process simulation and computer fluid dynamics) and process analytical technology (PAT) for seamless process development are emphasized. The work concludes with an outlook on future applications of refolding and highlights open research inquiries.
Collapse
Affiliation(s)
- Luisa Buscajoni
- Boehringer-Ingelheim RCV GmbH & Co KG, Biopharma Austria, Process Science Downstream Development, Dr. Boehringer-Gasse 5- 11, 1120 Vienna, Austria.
| | - Michael C Martinetz
- Boehringer-Ingelheim RCV GmbH & Co KG, Biopharma Austria, Process Science Downstream Development, Dr. Boehringer-Gasse 5- 11, 1120 Vienna, Austria.
| | - Matthias Berkemeyer
- Boehringer-Ingelheim RCV GmbH & Co KG, Biopharma Austria, Process Science Downstream Development, Dr. Boehringer-Gasse 5- 11, 1120 Vienna, Austria.
| | - Cécile Brocard
- Boehringer-Ingelheim RCV GmbH & Co KG, Biopharma Austria, Process Science Downstream Development, Dr. Boehringer-Gasse 5- 11, 1120 Vienna, Austria.
| |
Collapse
|
6
|
Kang MS, Kong TWS, Khoo JYX, Loh TP. Recent developments in chemical conjugation strategies targeting native amino acids in proteins and their applications in antibody-drug conjugates. Chem Sci 2021; 12:13613-13647. [PMID: 34760149 PMCID: PMC8549674 DOI: 10.1039/d1sc02973h] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022] Open
Abstract
Many fields in chemical biology and synthetic biology require effective bioconjugation methods to achieve their desired functions and activities. Among such biomolecule conjugates, antibody-drug conjugates (ADCs) need a linker that provides a stable linkage between cytotoxic drugs and antibodies, whilst conjugating in a biologically benign, fast and selective fashion. This review focuses on how the development of novel organic synthesis can solve the problems of traditional linker technology. The review shall introduce and analyse the current developments in the modification of native amino acids on peptides or proteins and their applicability to ADC linker. Thereafter, the review shall discuss in detail each endogenous amino acid's intrinsic reactivity and selectivity aspects, and address the research effort to construct an ADC using each conjugation method.
Collapse
Affiliation(s)
- Min Sun Kang
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Theresa Wai See Kong
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Joycelyn Yi Xin Khoo
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| | - Teck-Peng Loh
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University 21 Nanyang Link 637371 Singapore
| |
Collapse
|
7
|
A Single-Domain Antibody-Based Anti-PSMA Recombinant Immunotoxin Exhibits Specificity and Efficacy for Prostate Cancer Therapy. Int J Mol Sci 2021; 22:ijms22115501. [PMID: 34071152 PMCID: PMC8197099 DOI: 10.3390/ijms22115501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa) is the second most common cancer in men, causing more than 300,000 deaths every year worldwide. Due to their superior cell-killing ability and the relative simplicity of their preparation, immunotoxin molecules have great potential in the clinical treatment of cancer, and several such molecules have been approved for clinical application. In this study, we adopted a relatively simple strategy based on a single-domain antibody (sdAb) and an improved Pseudomonas exotoxin A (PE) toxin (PE24X7) to prepare a safer immunotoxin against prostate-specific membrane antigen (PSMA) for PCa treatment. The designed anti-PSMA immunotoxin, JVM-PE24X7, was conveniently prepared in its soluble form in an Escherichia coli (E. coli) system, avoiding the complex renaturation process needed for immunotoxin preparation by the conventional strategy. The product was very stable and showed a very strong ability to bind the PSMA receptor. Cytotoxicity assays showed that this molecule at a very low concentration could kill PSMA-positive PCa cells, with an EC50 value (concentration at which the cell viability decreased by 50%) of 15.3 pM against PSMA-positive LNCaP cells. Moreover, this molecule showed very good killing selectivity between PSMA-positive and PSMA-negative cells, with a selection ratio of more than 300-fold. Animal studies showed that this molecule at a very low dosage (5 × 0.5 mg/kg once every three days) completely inhibited the growth of PCa tumors, and the maximum tolerable dose (MTD) was more than 15 mg/kg, indicating its very potent tumor-treatment ability and a wide therapeutic window. Use of the new PE toxin, PE24X7, as the effector moiety significantly reduced off-target toxicity and improved the therapeutic window of the immunotoxin. The above results demonstrate that the designed anti-PSMA immunotoxin, JVM-PE24X7, has good application value for the treatment of PCa.
Collapse
|
8
|
Li Q, Li W, Xu K, Xing Y, Ding Y, Jing Z, Wang X, Hong Z. Preclinical evaluation of a novel anti-mesothelin immunotoxin based on a single domain antibody as the targeting ligand. Int J Pharm 2021; 602:120647. [PMID: 33915185 DOI: 10.1016/j.ijpharm.2021.120647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/21/2021] [Accepted: 04/22/2021] [Indexed: 01/10/2023]
Abstract
Pancreatic cancer, as one of the most aggressive and lethal malignancies in the world, is lack of effective treatment. Constructing immunotoxin molecules to target the mesothelin (MSLN) receptor is a potential therapeutic strategy for pancreatic cancer and other related malignant tumors, with some molecules being tested in clinical trials. However, currently, there are still some limitations in its applications, such as the difficulty of the preparation of drug molecules, the limited effectiveness of drugs, and the inadequacy of drug safety and immunogenicity. In this study, we constructed a novel type of anti-MSLN immunotoxin, A1-PE24X7, in which a single domain antibody (sdAb) molecule was used as the target ligand and an improved PE24X7 toxin with reduced off-target toxicity and immunogenicity was used as the effector. Unlike conventional immunotoxins, the designed A1-PE24X7 could be easily expressed in the E. coli system in the form of a soluble protein with a good yield (15--20 mg/L), avoiding the complex process of denaturation and refolding of inclusion bodies, and it can be conveniently stored in PBS solution for more than 7 days at 4 °C, showing high storage stability. Cell-based experiments showed that A1-PE24X7 entered MSLN-expressing tumor cells in a receptor-mediated manner and killed these cells with an EC50 in the low nanomolar range (0.13 nM against NCI-N87 cells and 0.79 nM against AsPC-1 cells) and it showed ideal selectivity for the MSLN receptor (>100 nM against receptor negative PC3 cells). In animal-based experiments, A1-PE24X7 had tumor enrichment ability in relation to MSLN-positive tumors and showed strong tumor killing and inhibition in mouse models of pancreatic cancer and gastric cancer. Five injections of 3.0 mg/kg A1-PE24X7 significantly reduced the tumor volume of gastric NCI-N87 cancer and also significantly inhibited the growth of pancreatic AsPC-1 cancer. In addition, the maximum tolerable dosage (MSD) of A1-PE24X7 to mice was higher than 15 mg/kg, showing that A1-PE24X7 has a relatively broad therapeutic window. These preclinical results indicate that this strategy has good potential for application to the treatment of pancreatic cancer and other tumors with high MSLN expression.
Collapse
Affiliation(s)
- Qiyu Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Wenjing Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Keyuan Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Yutong Xing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Yu Ding
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Zhe Jing
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Xi Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
9
|
Pauk JN, Raju Palanisamy J, Kager J, Koczka K, Berghammer G, Herwig C, Veiter L. Advances in monitoring and control of refolding kinetics combining PAT and modeling. Appl Microbiol Biotechnol 2021; 105:2243-2260. [PMID: 33598720 PMCID: PMC7954745 DOI: 10.1007/s00253-021-11151-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 12/21/2022]
Abstract
Overexpression of recombinant proteins in Escherichia coli results in misfolded and non-active protein aggregates in the cytoplasm, so-called inclusion bodies (IB). In recent years, a change in the mindset regarding IBs could be observed: IBs are no longer considered an unwanted waste product, but a valid alternative to produce a product with high yield, purity, and stability in short process times. However, solubilization of IBs and subsequent refolding is necessary to obtain a correctly folded and active product. This protein refolding process is a crucial downstream unit operation-commonly done as a dilution in batch or fed-batch mode. Drawbacks of the state-of-the-art include the following: the large volume of buffers and capacities of refolding tanks, issues with uniform mixing, challenging analytics at low protein concentrations, reaction kinetics in non-usable aggregates, and generally low re-folding yields. There is no generic platform procedure available and a lack of robust control strategies. The introduction of Quality by Design (QbD) is the method-of-choice to provide a controlled and reproducible refolding environment. However, reliable online monitoring techniques to describe the refolding kinetics in real-time are scarce. In our view, only monitoring and control of re-folding kinetics can ensure a productive, scalable, and versatile platform technology for re-folding processes. For this review, we screened the current literature for a combination of online process analytical technology (PAT) and modeling techniques to ensure a controlled refolding process. Based on our research, we propose an integrated approach based on the idea that all aspects that cannot be monitored directly are estimated via digital twins and used in real-time for process control. KEY POINTS: • Monitoring and a thorough understanding of refolding kinetics are essential for model-based control of refolding processes. • The introduction of Quality by Design combining Process Analytical Technology and modeling ensures a robust platform for inclusion body refolding.
Collapse
Affiliation(s)
- Jan Niklas Pauk
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, Vienna University of Technology, Gumpendorferstrasse 1a/166, 1060, Vienna, Austria
- Competence Center CHASE GmbH, Altenbergerstraße 69, 4040, Linz, Austria
| | - Janani Raju Palanisamy
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, Vienna University of Technology, Gumpendorferstrasse 1a/166, 1060, Vienna, Austria
| | - Julian Kager
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, Vienna University of Technology, Gumpendorferstrasse 1a/166, 1060, Vienna, Austria
| | - Krisztina Koczka
- Bilfinger Industrietechnik Salzburg GmbH, Mooslackengasse 17, 1190, Vienna, Austria
| | - Gerald Berghammer
- Bilfinger Industrietechnik Salzburg GmbH, Mooslackengasse 17, 1190, Vienna, Austria
| | - Christoph Herwig
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, Vienna University of Technology, Gumpendorferstrasse 1a/166, 1060, Vienna, Austria.
| | - Lukas Veiter
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, Vienna University of Technology, Gumpendorferstrasse 1a/166, 1060, Vienna, Austria
- Competence Center CHASE GmbH, Altenbergerstraße 69, 4040, Linz, Austria
| |
Collapse
|
10
|
Lu X, Lin S, De Mel N, Parupudi A, Delmar J, Pandey M, Wang X, Wang J. Deamidation in Moxetumomab Pasudotox Leading to Conformational Change and Immunotoxin Activity Loss. J Pharm Sci 2020; 109:2676-2683. [PMID: 32534028 DOI: 10.1016/j.xphs.2020.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/14/2020] [Accepted: 06/01/2020] [Indexed: 10/24/2022]
Abstract
Asparagine (Asn) deamidation is a common posttranslational modification in which Asn is converted to aspartic acid or isoaspartic acid. By introducing a negative charge, deamidation could potentially impact the binding interface and biological activities of protein therapeutics. We identified a deamidation variant in moxetumomab pasudotox, an immunotoxin Fv fusion protein drug derived from a 38-kDa truncated Pseudomonas exotoxin A (PE38) for the treatment of hairy-cell leukemia. Although the deamidation site, Asn-358, was outside of the binding interface, the modification had a significant impact on the biological activity of moxetumomab pasudotox. Surprisingly, the variant eluted earlier than its unmodified form on anion exchange chromatography, which often leads to the conclusion that it has a higher positive charge. Here we describe the characterization of the deamidation variant with differential scanning calorimetry and hydrogen-deuterium exchange mass spectrometry, which revealed that the Asn-358 deamidation caused the conformational changes in the catalytic domain of the PE38 region. These results provide an explanation for why the deamidation affected the biological activity of moxetumomab pasudotox and suggest the approach that can be used for process control to ensure product quality and process consistency.
Collapse
Affiliation(s)
- Xiaojun Lu
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, Maryland 20878
| | - Shihua Lin
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, Maryland 20878
| | - Niluka De Mel
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, Maryland 20878
| | - Arun Parupudi
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, Maryland 20878
| | - Jared Delmar
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, Maryland 20878
| | - Madhu Pandey
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, Maryland 20878
| | - Xiangyang Wang
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, Maryland 20878
| | - Jihong Wang
- Analytical Sciences, Biopharmaceutical Development, AstraZeneca, One MedImmune Way, Gaithersburg, Maryland 20878.
| |
Collapse
|
11
|
Cao L, Li Q, Tong Z, Xing Y, Xu K, Yijia Wang J, Li W, Zhao J, Zhao L, Hong Z. HER2-specific immunotoxins constructed based on single-domain antibodies and the improved toxin PE24X7. Int J Pharm 2020; 574:118939. [DOI: 10.1016/j.ijpharm.2019.118939] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/11/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023]
|
12
|
Xi G, Esfandiary R, Sacramento CB, Jouihan H, Sharma A, Roth R, Linke T. Refolding and purification of cGMP-grade recombinant human neurturin from Escherichia coli inclusion bodies. Protein Expr Purif 2019; 168:105552. [PMID: 31866372 DOI: 10.1016/j.pep.2019.105552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/18/2019] [Indexed: 10/25/2022]
Abstract
Neurturin is a potent neurotrophic factor that has been investigated as a potential therapeutic agent for the treatment of neurodegenerative diseases, including Parkinson's disease, and, more recently, for the treatment of type II diabetes. However, purification of neurturin for clinical applications has been hampered by its low solubility in aqueous solutions. Here we describe the development of a scalable manufacturing process for recombinant neurturin from E. coli. inclusion bodies. Neurturin was refolded from solubilized inclusion bodies by fed-batch dilution refolding with a titer of 90 mg per liter refold and a refold yield of 89%. A two-step purification process using cation exchange and hydrophobic interaction chromatography, followed by formulation using tangential flow filtration resulted in an overall process yield of about 56 mg purified neurturin per liter refold. Solubility of neurturin during the purification process was maintained by the addition of 15% (w/v) glycerol to all buffers. For clinical applications and parenteral administration glycerol was replaced by 15% (w/v) sulfobutyl ether-beta-cyclodextrin (i.e. Captisol) in the drug substance formulation buffer. The final purified product had low or undetectable levels of product-related impurities and concentrations of process-related contaminants such as host cell proteins, host cell DNA, endotoxins and Triton X-100 were reduced more than 10,000-fold or below the limit of detection. Bioactivity of purified recombinant neurturin was demonstrated in a cell-based assay by activation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Guoling Xi
- Department of Purification Process Sciences, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Reza Esfandiary
- Department of Dosage Form Design and Development, AstraZeneca, Gaithersburg, MD, 20878, USA
| | | | - Hani Jouihan
- Department of Cardiovascular and Metabolic Diseases, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Arun Sharma
- Department of Cardiovascular and Metabolic Diseases, AstraZeneca, Gaithersburg, MD, 20878, USA
| | - Robert Roth
- Discovery Biology, Discovery Sciences, AstraZeneca, Gothenburg, 43183, Sweden
| | - Thomas Linke
- Department of Purification Process Sciences, AstraZeneca, Gaithersburg, MD, 20878, USA.
| |
Collapse
|
13
|
Critical Issues in the Development of Immunotoxins for Anticancer Therapy. J Pharm Sci 2019; 109:104-115. [PMID: 31669121 DOI: 10.1016/j.xphs.2019.10.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
Abstract
Immunotoxins (ITs) are attractive anticancer modalities aimed at cancer-specific delivery of highly potent cytotoxic protein toxins. An IT consists of a targeting domain (an antibody, cytokine, or another cell-binding protein) chemically conjugated or recombinantly fused to a highly cytotoxic payload (a bacterial and plant toxin or human cytotoxic protein). The mode of action of ITs is killing designated cancer cells through the effector function of toxins in the cytosol after cellular internalization via the targeted cell-specific receptor-mediated endocytosis. Although numerous ITs of diverse structures have been tested in the past decades, only 3 ITs-denileukin diftitox, tagraxofusp, and moxetumomab pasudotox-have been clinically approved for treating hematological cancers. No ITs against solid tumors have been approved for clinical use. In this review, we discuss critical research and development issues associated with ITs that limit their clinical success as well as strategies to overcome these obstacles. The issues include off-target and on-target toxicities, immunogenicity, human cytotoxic proteins, antigen target selection, cytosolic delivery efficacy, solid-tumor targeting, and developability. To realize the therapeutic promise of ITs, novel strategies for safe and effective cytosolic delivery into designated tumors, including solid tumors, are urgently needed.
Collapse
|
14
|
Bokori-Brown M, Metz J, Petrov PG, Mussai F, De Santo C, Smart NJ, Saunders S, Knight B, Pastan I, Titball RW, Winlove CP. Interactions Between Pseudomonas Immunotoxins and the Plasma Membrane: Implications for CAT-8015 Immunotoxin Therapy. Front Oncol 2018; 8:553. [PMID: 30538953 PMCID: PMC6277520 DOI: 10.3389/fonc.2018.00553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/08/2018] [Indexed: 11/13/2022] Open
Abstract
Acute Lymphoblastic Leukemia (ALL) remains the most frequent cause of cancer-related mortality in children and novel therapies are needed for the treatment of relapsed/refractory childhood ALL. One approach is the targeting of ALL blasts with the Pseudomonas immunotoxin CAT-8015. Although CAT-8015 has potent anti-leukemia activity, with a 32% objective response rate in a phase 1 study of childhood ALL, haemolytic-uremic syndrome (HUS) and vascular leak syndrome (VLS), major dose-limiting toxicities, have limited the use of this therapeutic approach in children. Investigations into the pathogenesis of CAT-8015-induced HUS/VLS are hindered by the lack of an adequate model system that replicates clinical manifestations, but damage to vascular endothelial cells (ECs) and blood cells are believed to be major initiating factors in both syndromes. Since there is little evidence that murine models replicate human HUS/VLS, and CAT-8015-induced HUS/VLS predominantly affects children, we developed human models and used novel methodologies to investigate CAT-8015 interactions with red blood cells (RBCs) from pediatric ALL patients and ECs of excised human mesenteric arteries. We provide evidence that CAT-8015 directly interacts with RBCs, mediated by Pseudomonas toxin. We also show correlation between the electrical properties of the RBC membrane and RBC susceptibility to CAT-8015-induced lysis, which may have clinical implication. Finally, we provide evidence that CAT-8015 is directly cytototoxic to ECs of excised human mesenteric arteries. In conclusion, the human models we developed constitutes the first, and very important, step in understanding the origins of HUS/VLS in immunotoxin therapy and will allow further investigations of HUS/VLS pathogenesis.
Collapse
Affiliation(s)
- Monika Bokori-Brown
- College of Life and Environmental Sciences, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Jeremy Metz
- College of Life and Environmental Sciences, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Peter G. Petrov
- College of Engineering, Mathematics and Physical Sciences, Department of Physics and Astronomy, University of Exeter, Exeter, United Kingdom
| | - Francis Mussai
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Carmela De Santo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Neil J. Smart
- Exeter Surgical Health Services Research Unit, Royal Devon and Exeter Hospital, Exeter, United Kingdom
| | - Sarah Saunders
- Histopathology Department, Royal Devon and Exeter Hospital, Exeter, United Kingdom
| | - Bridget Knight
- National Institute for Health Research Exeter Clinical Research Facility, Royal Devon and Exeter National Health Service Foundation Trust, Exeter, United Kingdom
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Richard W. Titball
- College of Life and Environmental Sciences, School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - C. Peter Winlove
- College of Engineering, Mathematics and Physical Sciences, Department of Physics and Astronomy, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
15
|
Humer D, Spadiut O. Wanted: more monitoring and control during inclusion body processing. World J Microbiol Biotechnol 2018; 34:158. [PMID: 30341583 PMCID: PMC6208753 DOI: 10.1007/s11274-018-2541-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/11/2018] [Indexed: 01/20/2023]
Abstract
Inclusion bodies (IBs) are insoluble aggregates of misfolded protein in Escherichia coli. Against the outdated belief that the production of IBs should be avoided during recombinant protein production, quite a number of recombinant products are currently produced as IBs, which are then processed to give correctly folded and soluble product. However, this processing is quite cumbersome comprising IB wash, IB solubilization and refolding. To date, IB processing often happens rather uncontrolled and relies on empiricism rather than sound process understanding. In this mini review we describe current efforts to introduce more monitoring and control in IB processes, focusing on the refolding step, and thus generate process understanding and knowledge.
Collapse
Affiliation(s)
- Diana Humer
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorfer Straße 1a, 1060, Vienna, Austria
| | - Oliver Spadiut
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorfer Straße 1a, 1060, Vienna, Austria.
| |
Collapse
|
16
|
Kinjyo I, Matlawska-Wasowska K, Chen X, Monks NR, Burke P, Winter SS, Wilson BS. Characterization of the anti-CD22 targeted therapy, moxetumomab pasudotox, for B-cell precursor acute lymphoblastic leukemia. Pediatr Blood Cancer 2017; 64:10.1002/pbc.26604. [PMID: 28449314 PMCID: PMC7501879 DOI: 10.1002/pbc.26604] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/15/2017] [Accepted: 03/18/2017] [Indexed: 11/10/2022]
Abstract
Moxetumomab pasudotox is a second-generation recombinant immunotoxin against CD22 on B-cell lineages. Antileukemic activity has been demonstrated in children with chemotherapy-refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL), with variable responses. Here, we report in vitro and in vivo evaluation of moxetumomab pasudotox treatment of human cell lines and patient-derived cells as a preliminary study to understand characteristics of sensitivity to treatment. Binding, internalization, and apoptosis were evaluated using fluorescently tagged moxetumomab pasudotox. Studies in NOD-scid IL2Rgnull mice showed a modest survival benefit in mice engrafted with 697 cells but not in NALM6 or the two patient-derived xenograft models.
Collapse
Affiliation(s)
- Ichiko Kinjyo
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ksenia Matlawska-Wasowska
- Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico,Division of Hematology/Oncology, Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Xiaoru Chen
- Translational Medicine Oncology, MedImmune, Gaithersburg, Maryland
| | - Noel R. Monks
- Oncology Research, MedImmune, Gaithersburg, Maryland
| | - Patricia Burke
- Translational Medicine Oncology, MedImmune, Gaithersburg, Maryland
| | - Stuart S. Winter
- Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico,Division of Hematology/Oncology, Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Bridget S. Wilson
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico,Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
17
|
Wayne AS, Shah NN, Bhojwani D, Silverman LB, Whitlock JA, Stetler-Stevenson M, Sun W, Liang M, Yang J, Kreitman RJ, Lanasa MC, Pastan I. Phase 1 study of the anti-CD22 immunotoxin moxetumomab pasudotox for childhood acute lymphoblastic leukemia. Blood 2017; 130:1620-1627. [PMID: 28983018 PMCID: PMC5630009 DOI: 10.1182/blood-2017-02-749101] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/27/2017] [Indexed: 02/08/2023] Open
Abstract
Novel therapies are needed to overcome chemotherapy resistance for children with relapsed/refractory acute lymphoblastic leukemia (ALL). Moxetumomab pasudotox is a recombinant anti-CD22 immunotoxin. A multicenter phase 1 study was conducted to determine the maximum-tolerated cumulative dose (MTCD) and evaluate safety, activity, pharmacokinetics, and immunogenicity of moxetumomab pasudotox in children, adolescents, and young adults with ALL (N = 55). Moxetumomab pasudotox was administered as a 30-minute IV infusion at doses of 5 to 50 µg/kg every other day for 6 (cohorts A and B) or 10 (cohort C) doses in 21-day cycles. Cohorts B and C received dexamethasone prophylaxis against capillary leak syndrome (CLS). The most common treatment-related adverse events were reversible weight gain, hepatic transaminase elevation, and hypoalbuminemia. Dose-limiting CLS occurred in 2 of 4 patients receiving 30 µg/kg of moxetumomab pasudotox every other day for 6 doses. Incorporation of dexamethasone prevented further dose-limiting CLS. Six of 14 patients receiving 50 µg/kg of moxetumomab pasudotox for 10 doses developed hemolytic uremic syndrome (HUS), thrombotic microangiopathy (TMA), or HUS-like events, exceeding the MTCD. Treatment expansion at 40 µg/kg for 10 doses (n = 11) exceeded the MTCD because of 2 HUS/TMA/HUS-like events. Dose level 6B (ie, 50 µg/kg × 6 doses) was the MTCD, selected as the recommended phase 2 dose. Among 47 evaluable patients, an objective response rate of 32% was observed, including 11 (23%) composite complete responses, 5 of which were minimal residual disease negative by flow cytometry. Moxetumomab pasudotox showed a manageable safety profile and evidence of activity in relapsed or refractory childhood ALL. This trial was registered at www.clinicaltrials.gov as #NCT00659425.
Collapse
Affiliation(s)
- Alan S Wayne
- Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
- Laboratory of Molecular Biology, CCR, NCI, NIH, Bethesda, MD
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD
| | - Deepa Bhojwani
- Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
- St Jude Children's Research Hospital, Memphis, TN
| | - Lewis B Silverman
- Pediatric Hematologic Malignancies Center, Dana-Farber Cancer Institute/Boston Children's Hospital, Boston, MA
| | - James A Whitlock
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | - Weili Sun
- Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | | | | | | | - Ira Pastan
- Laboratory of Molecular Biology, CCR, NCI, NIH, Bethesda, MD
| |
Collapse
|
18
|
Mousavi SB, Fazeli A, Shojaosadati SA, Fazeli MR, Hashemi-Najafabadi S. Purification and efficient refolding process for recombinant tissue-type plasminogen activator derivative (reteplase) using glycerol and Tranexamic acid. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.11.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
19
|
Pabst TM, Wendeler M, Wang X, Bezemer S, Hermans P, Hunter AK. Camelid V H H affinity ligands enable separation of closely related biopharmaceuticals. Biotechnol J 2016; 12. [PMID: 27677057 PMCID: PMC5333455 DOI: 10.1002/biot.201600357] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/01/2016] [Accepted: 09/27/2016] [Indexed: 01/15/2023]
Abstract
Interest in new and diverse classes of molecules such as recombinant toxins, enzymes, and blood factors continues to grow for use a biotherapeutics. Compared to monoclonal antibodies, these novel drugs typically lack a commercially available affinity chromatography option, which leads to greater process complexity, longer development timelines, and poor platformability. To date, for both monoclonal antibodies and novel molecules, affinity chromatography has been mostly reserved for separation of process‐related impurities such as host cell proteins and DNA. Reports of affinity purification of closely related product variants and modified forms are much rarer. In this work we describe custom affinity chromatography development using camelid VHH antibody fragments as "tunable" immunoaffinity ligands for separation of product‐related impurities. One example demonstrates high selectivity for a recombinant immunotoxin where no binding was observed for an undesired deamidated species. Also discussed is affinity purification of a coagulation factor through specific recognition of the gamma‐carboxylglutamic acid domain.
Collapse
Affiliation(s)
- Timothy M Pabst
- MedImmune, Department of Purification Process Sciences, Gaithersburg, MD, USA
| | - Michaela Wendeler
- MedImmune, Department of Purification Process Sciences, Gaithersburg, MD, USA
| | - Xiangyang Wang
- MedImmune, Department of Purification Process Sciences, Gaithersburg, MD, USA
| | | | - Pim Hermans
- Thermo Fisher Scientific, Naarden, The Netherlands
| | - Alan K Hunter
- MedImmune, Department of Purification Process Sciences, Gaithersburg, MD, USA
| |
Collapse
|
20
|
Engineering batch and pulse refolding with transition of aggregation kinetics: An investigation using green fluorescent protein (GFP). Chem Eng Sci 2015. [DOI: 10.1016/j.ces.2015.03.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|