1
|
Nawab DH. Vaccinal antibodies: Fc antibody engineering to improve the antiviral antibody response and induce vaccine-like effects. Hum Vaccin Immunother 2021; 17:5532-5545. [PMID: 34844516 PMCID: PMC8903937 DOI: 10.1080/21645515.2021.1985891] [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: 04/25/2021] [Accepted: 09/21/2021] [Indexed: 10/19/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic highlights the urgent clinical need for efficient virus therapies and vaccines. Although the functional importance of antibodies is indisputable in viral infections, there are still significant unmet needs that require vast improvements in antibody-based therapeutics. The IgG Fc domain can be engineered to produce antibodies with tailored and potent responses that will meet these clinical demands. Engaging Fc receptors (FcRs) to perform effector functions as cytotoxicity, phagocytosis, complement activation, intracellular neutralization and controlling antibody persistence. Furthermore, it produces vaccine-like effects by activating signals to stimulate T-cell responses, have proven to be required for protection, as neutralization alone does not off the full protection capacity of antibodies. This review highlights antiviral Fc functions and FcRs' contributions in linking innate and adaptive immunity against viral threats. Moreover, it provides the latest Fc engineering strategies to improve the safety and efficacy of human antiviral antibodies and vaccines.
Collapse
Affiliation(s)
- Dhuha H Nawab
- Pharmacy Department, Ministry of Health, Saudi Arabia
| |
Collapse
|
2
|
Expression and purification of a novel single-chain diabody (scDb-hERG1/β1) from Pichia pastoris transformants. Protein Expr Purif 2021; 184:105879. [PMID: 33826963 DOI: 10.1016/j.pep.2021.105879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/01/2021] [Accepted: 03/31/2021] [Indexed: 01/07/2023]
Abstract
In the last decades, protein engineering has developed particularly in biotechnology and pharmaceutical field. In particular, the engineered antibody subclass has arisen. The single chain diabody format (scDb), conjugating small size with antigen specificity, offers versatility representing a gold standard for a variety of applications, spacing from research to diagnostics and therapy. Along with such advantages, comes the challenge of optimizing their production, improving expression systems, purification procedures and stability. All such parameters are detrimental for protein production in general and above all for recombinant antibody expression, which has to be fine-tuned, choosing a proper protein-expression host and adjusting expression protocols accordingly. In the present paper, we present data regarding the production and purification of a single chain diabody directed against the macromolecular complex hERG1/β1 integrin. We focus on the expression of clones deriving from the transformation of Pichia pastoris yeast cells. In particular, we compare two different clones arose from two separate transformation processes, demonstrating that both are suitable for proper protein expression. Moreover, we have set up an expression protocol and compared the yields obtained using two purification machines: Akta Pure and Akta Start, with a positive outcome.
Collapse
|
3
|
Lardinois OM, Deterding LJ, Hess JJ, Poulton CJ, Henderson CD, Jennette JC, Nachman PH, Falk RJ. Immunoglobulins G from patients with ANCA-associated vasculitis are atypically glycosylated in both the Fc and Fab regions and the relation to disease activity. PLoS One 2019; 14:e0213215. [PMID: 30818380 PMCID: PMC6395067 DOI: 10.1371/journal.pone.0213215] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/15/2019] [Indexed: 02/07/2023] Open
Abstract
Background Anti-neutrophil cytoplasmic autoantibodies (ANCA) directed against myeloperoxidase (MPO) and proteinase 3 (PR3) are pathogenic in ANCA-associated vasculitis (AAV). The respective role of IgG Fc and Fab glycosylation in mediating ANCA pathogenicity is incompletely understood. Herein we investigate in detail the changes in Fc and Fab glycosylation in MPO-ANCA and Pr3-ANCA and examine the association of glycosylation aberrancies with disease activity. Methodology Total IgG was isolated from serum or plasma of a cohort of 30 patients with AAV (14 MPO-ANCA; 16 PR3-ANCA), and 19 healthy control subjects. Anti-MPO specific IgG was affinity-purified from plasma of an additional cohort of 18 MPO-ANCA patients undergoing plasmapheresis. We used lectin binding assays, liquid chromatography, and mass spectrometry-based methods to analyze Fc and Fab glycosylation, the degree of sialylation of Fc and Fab fragments and to determine the exact localization of N-glycans on Fc and Fab fragments. Principal findings IgG1 Fc glycosylation of total IgG was significantly reduced in patients with active AAV compared to controls. Clinical remission was associated with complete glycan normalization for PR3-ANCA patients but not for MPO-ANCA patients. Fc-glycosylation of anti-MPO specific IgG was similar to total IgG purified from plasma. A major fraction of anti-MPO specific IgG harbor extensive glycosylation within the variable domain on the Fab portion. Conclusions/Significance Significant differences exist between MPO and PR3-ANCA regarding the changes in amounts and types of glycans on Fc fragment and the association with disease activity. These differences may contribute to significant clinical difference in the disease course observed between the two diseases.
Collapse
Affiliation(s)
- Olivier M. Lardinois
- UNC Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Mass Spectrometry Research and Support Group, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| | - Leesa J. Deterding
- Mass Spectrometry Research and Support Group, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, United States of America
| | - Jacob J. Hess
- UNC Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Caroline J. Poulton
- UNC Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Candace D. Henderson
- UNC Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - J. Charles Jennette
- Department of Pathology and Laboratory of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Patrick H. Nachman
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ronald J. Falk
- UNC Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
4
|
Buettner MJ, Shah SR, Saeui CT, Ariss R, Yarema KJ. Improving Immunotherapy Through Glycodesign. Front Immunol 2018; 9:2485. [PMID: 30450094 PMCID: PMC6224361 DOI: 10.3389/fimmu.2018.02485] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/08/2018] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy is revolutionizing health care, with the majority of high impact "drugs" approved in the past decade falling into this category of therapy. Despite considerable success, glycosylation-a key design parameter that ensures safety, optimizes biological response, and influences the pharmacokinetic properties of an immunotherapeutic-has slowed the development of this class of drugs in the past and remains challenging at present. This article describes how optimizing glycosylation through a variety of glycoengineering strategies provides enticing opportunities to not only avoid past pitfalls, but also to substantially improve immunotherapies including antibodies and recombinant proteins, and cell-based therapies. We cover design principles important for early stage pre-clinical development and also discuss how various glycoengineering strategies can augment the biomanufacturing process to ensure the overall effectiveness of immunotherapeutics.
Collapse
Affiliation(s)
- Matthew J Buettner
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Sagar R Shah
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Christopher T Saeui
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States.,Pharmacology/Toxicology Branch I, Division of Clinical Evaluation and Pharmacology/Toxicology, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, MD, United States
| | - Ryan Ariss
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kevin J Yarema
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
5
|
Roberts JT, Barb AW. A single amino acid distorts the Fc γ receptor IIIb/CD16b structure upon binding immunoglobulin G1 and reduces affinity relative to CD16a. J Biol Chem 2018; 293:19899-19908. [PMID: 30361439 DOI: 10.1074/jbc.ra118.005273] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/15/2018] [Indexed: 12/31/2022] Open
Abstract
Therapeutic mAbs engage Fc γ receptor III (CD16) to elicit a protective cell-mediated response and destroy the target tissue. Newer drugs designed to bind CD16a with increased affinity surprisingly also elicit protective CD16b-mediated responses. However, it is unclear why IgG binds CD16a with more than 10-fold higher affinity than CD16b even though these receptors share more than 97% identity. Here we identified one residue, Gly-129, that contributes to the greater IgG binding affinity of CD16a. The CD16b variant D129G bound IgG1 Fc with 2-fold higher affinity than CD16a and with 90-fold higher affinity than the WT. Conversely, the binding affinity of CD16a-G129D was decreased 128-fold relative to WT CD16a and comparably to that of WT CD16b. The interaction of IgG1 Fc with CD16a, but not with CD16b, is known to be sensitive to the composition of the asparagine-linked carbohydrates (N-glycans) attached to the receptor. CD16a and CD16b-D129G displaying minimally processed oligomannose N-glycans bound to IgG1 Fc with about 5.2-fold increased affinity compared with variants with highly processed complex-type N-glycans. CD16b and the CD16a-G129D variant exhibited a smaller 1.9-fold affinity increase with oligomannose N-glycans. A model of glycosylated CD16b bound to IgG1 Fc determined to 2.2 Å resolution combined with a 250-ns all-atom molecular dynamics simulation showed that the larger Asp-129 residue deformed the Fc-binding surface. These results reveal how Asp-129 in CD16b affects its binding affinity for IgG1 Fc and suggest that antibodies engineered to engage CD16b with high affinity must accommodate the Asp-129 side chain.
Collapse
Affiliation(s)
- Jacob T Roberts
- From the Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, Iowa 50011
| | - Adam W Barb
- From the Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, Iowa 50011
| |
Collapse
|
6
|
Veillon L, Fakih C, Abou-El-Hassan H, Kobeissy F, Mechref Y. Glycosylation Changes in Brain Cancer. ACS Chem Neurosci 2018; 9:51-72. [PMID: 28982002 DOI: 10.1021/acschemneuro.7b00271] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Protein glycosylation is a posttranslational modification that affects more than half of all known proteins. Glycans covalently bound to biomolecules modulate their functions by both direct interactions, such as the recognition of glycan structures by binding partners, and indirect mechanisms that contribute to the control of protein conformation, stability, and turnover. The focus of this Review is the discussion of aberrant glycosylation related to brain cancer. Altered sialylation and fucosylation of N- and O-glycans play a role in the development and progression of brain cancer. Additionally, aberrant O-glycan expression has been implicated in brain cancer. This Review also addresses the clinical potential and applications of aberrant glycosylation for the detection and treatment of brain cancer. The viable roles glycans may play in the development of brain cancer therapeutics are addressed as well as cancer-glycoproteomics and personalized medicine. Glycoprotein alterations are considered as a hallmark of cancer while high expression in body fluids represents an opportunity for cancer assessment.
Collapse
Affiliation(s)
- Lucas Veillon
- Department
of Chemistry and Biochemistry, Texas Tech University, Lubbock Texas 79409, United States
| | - Christina Fakih
- Department
of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hadi Abou-El-Hassan
- Department
of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Firas Kobeissy
- Department
of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yehia Mechref
- Department
of Chemistry and Biochemistry, Texas Tech University, Lubbock Texas 79409, United States
| |
Collapse
|
7
|
Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs 2017; 9:182-212. [PMID: 28071970 PMCID: PMC5297537 DOI: 10.1080/19420862.2016.1268307] [Citation(s) in RCA: 605] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022] Open
Abstract
During the past two decades we have seen a phenomenal evolution of bispecific antibodies for therapeutic applications. The 'zoo' of bispecific antibodies is populated by many different species, comprising around 100 different formats, including small molecules composed solely of the antigen-binding sites of two antibodies, molecules with an IgG structure, and large complex molecules composed of different antigen-binding moieties often combined with dimerization modules. The application of sophisticated molecular design and genetic engineering has solved many of the technical problems associated with the formation of bispecific antibodies such as stability, solubility and other parameters that confer drug properties. These parameters may be summarized under the term 'developability'. In addition, different 'target product profiles', i.e., desired features of the bispecific antibody to be generated, mandates the need for access to a diverse panel of formats. These may vary in size, arrangement, valencies, flexibility and geometry of their binding modules, as well as in their distribution and pharmacokinetic properties. There is not 'one best format' for generating bispecific antibodies, and no single format is suitable for all, or even most of, the desired applications. Instead, the bispecific formats collectively serve as a valuable source of diversity that can be applied to the development of therapeutics for various indications. Here, a comprehensive overview of the different bispecific antibody formats is provided.
Collapse
Affiliation(s)
- Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Im Nonnenwald, Penzberg, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Nobelstraße, Stuttgart, Germany
| |
Collapse
|
8
|
Epp A, Sullivan KC, Herr AB, Strait RT. Immunoglobulin Glycosylation Effects in Allergy and Immunity. Curr Allergy Asthma Rep 2017; 16:79. [PMID: 27796794 DOI: 10.1007/s11882-016-0658-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The aim of this review will be to familiarize the reader with the general area of antibody (Ab) glycosylation and to summarize the known functional roles of glycosylation and how glycan structure can contribute to various disease states with emphasis on allergic disease. RECENT FINDINGS Both immunoglobulin (Ig) isotype and conserved Fc glycosylation sites often dictate the downstream activity of an Ab where complexity and degree of glycosylation contribute to its ability to bind Fc receptors (FcRs) and activate complement. Most information on the effects of glycosylation center on IgG in cancer therapy and autoimmunity. In cancer therapy, glycosylation modifications that enhance affinity for activating FcRs are utilized to facilitate immune-mediated tumor cell killing. In autoimmunity, disease severity has been linked to alterations in the presence, location, and composition of Fc glycans. Significantly less is understood about the role of glycosylation in the setting of allergy and asthma. However, recent data demonstrate that glycosylation of IgE at the asparagine-394 site of Cε3 is necessary for IgE interaction with the high affinity IgE receptor but, surprisingly, glycosylation has no effect on IgE interaction with its low-affinity lectin receptor, CD23. Variations in the specific glycoform may modulate the interaction of an Ig with its receptors. Significantly more is known about the functional effects of glycosylation of IgG than for other Ig isotypes. Thus, the role of glycosylation is much better understood in the areas of autoimmunity and cancer therapy, where IgG is the dominant isotype, than in the field of allergy, where IgE predominates. Further work is needed to fully understand the role of glycan variation in IgE and other Ig isotypes with regard to the inhibition or mediation of allergic disease.
Collapse
Affiliation(s)
- Alexandra Epp
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Kathryn C Sullivan
- Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Andrew B Herr
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Richard T Strait
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA. .,Division of Emergency Medicine, Cincinnati Children's Hospital, 3333 Burnet Ave, ML 2008, Cincinnati, OH, 45229, USA.
| |
Collapse
|
9
|
Hayes JM, Wormald MR, Rudd PM, Davey GP. Fc gamma receptors: glycobiology and therapeutic prospects. J Inflamm Res 2016; 9:209-219. [PMID: 27895507 PMCID: PMC5118039 DOI: 10.2147/jir.s121233] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Therapeutic antibodies hold great promise for the treatment of cancer and autoimmune diseases, and developments in antibody–drug conjugates and bispecific antibodies continue to enhance treatment options for patients. Immunoglobulin (Ig) G antibodies are proteins with complex modifications, which have a significant impact on their function. The most important of these modifications is glycosylation, the addition of conserved glycans to the antibody Fc region, which is critical for its interaction with the immune system and induction of effector activities such as antibody-dependent cell cytotoxicity, complement activation and phagocytosis. Communication of IgG antibodies with the immune system is controlled and mediated by Fc gamma receptors (FcγRs), membrane-bound proteins, which relay the information sensed and gathered by antibodies to the immune system. These receptors are also glycoproteins and provide a link between the innate and adaptive immune systems. Recent information suggests that this receptor glycan modification is also important for the interaction with antibodies and downstream immune response. In this study, the current knowledge on FcγR glycosylation is discussed, and some insight into its role and influence on the interaction properties with IgG, particularly in the context of biotherapeutics, is provided. For the purpose of this study, other Fc receptors such as FcαR, FcεR or FcRn are not discussed extensively, as IgG-based antibodies are currently the only therapeutic antibody-based products on the market. In addition, FcγRs as therapeutics and therapeutic targets are discussed, and insight into and comment on the therapeutic aspects of receptor glycosylation are provided.
Collapse
Affiliation(s)
- Jerrard M Hayes
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Mark R Wormald
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, UK
| | - Pauline M Rudd
- NIBRT Glycoscience Group, National Institute for Bioprocessing, Research and Training, Dublin, Ireland
| | - Gavin P Davey
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| |
Collapse
|
10
|
Konstantinus IN, Gamieldien H, Mkhize NN, Kriek JM, Passmore JAS. Comparing high-throughput methods to measure NK cell-mediated antibody dependent cellular cytotoxicity during HIV-infection. J Immunol Methods 2016; 434:46-52. [PMID: 27094485 DOI: 10.1016/j.jim.2016.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/23/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022]
Abstract
HIV-specific binding antibody responses, including those mediating antibody-dependent cellular cytotoxicity (ADCC), provided the best functional correlate of lower risk of infection in the RV144 HIV-1 vaccine clinical trial. The aim of this study was to compare two high-throughput flow cytometry based methods to measure HIV-specific ADCC responses, the GranToxilux and PanToxilux assays. Plasma from nine HIV-1 seropositive individuals was screened for binding antibody titres against HIV-1 subtype C gp120 by ELISA and western blot. Plasma from six HIV-negative individuals was included as controls. Both ADCC assays used subtype C gp120-coated CEM.NKRCCR5 cells as targets. The PanToxilux assay (which measured both granzyme B and caspase activity) measured higher levels of direct natural killer (NK) cell killing of K562 tumour cells than the GranToxilux assay (granzyme B alone; p<0.05). In ADCC assays in which NK cell killing was directed against gp120-coated CEM.NKRCCR5 cells in an antibody-dependent manner, plasma from HIV-positive individuals yielded significantly higher levels of ADCC activity than the HIV-negative controls. In contrast to direct killing, the GranToxilux assay measured similar levels of ADCC killing as the PanToxilux assay but had significantly lower background cytotoxicity against target cells coated with HIV negative serum. In conclusion, the PanToxilux assay was more sensitive for detecting direct NK cell killing of K562 cells than the GranToxilux assay, although the GranToxilux assay performed better at detecting HIV-specific ADCC activity, because of lower background cytotoxicity from HIV-negative serum. This is the first study to compare GranToxilux and PanToxilux ability to detect ADCC during HIV infection.
Collapse
Affiliation(s)
- Iyaloo N Konstantinus
- Institute of Infectious Diseases and Molecular Medicine, Division of Medical Virology, University of Cape Town, South Africa
| | - Hoyam Gamieldien
- Institute of Infectious Diseases and Molecular Medicine, Division of Medical Virology, University of Cape Town, South Africa
| | - Nonhlanhla N Mkhize
- National Institute for Communicable Diseases of the National Health Laboratory Services, South Africa
| | - Jean-Mari Kriek
- Institute of Infectious Diseases and Molecular Medicine, Division of Medical Virology, University of Cape Town, South Africa
| | - Jo-Ann S Passmore
- Institute of Infectious Diseases and Molecular Medicine, Division of Medical Virology, University of Cape Town, South Africa; National Health Laboratory Service, Cape Town, South Africa.
| |
Collapse
|
11
|
Pelegrin M, Naranjo-Gomez M, Piechaczyk M. Antiviral Monoclonal Antibodies: Can They Be More Than Simple Neutralizing Agents? Trends Microbiol 2016; 23:653-665. [PMID: 26433697 PMCID: PMC7127033 DOI: 10.1016/j.tim.2015.07.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/06/2015] [Accepted: 07/15/2015] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies (mAbs) are increasingly being considered as agents to fight severe viral diseases. So far, they have essentially been selected and used on the basis of their virus-neutralizing activity and/or cell-killing activity to blunt viral propagation via direct mechanisms. There is, however, accumulating evidence that they can also induce long-lasting protective antiviral immunity by recruiting the endogenous immune system of infected individuals during the period of immunotherapy. Exploiting this property may revolutionize antiviral mAb-based immunotherapies, with benefits for both patients and healthcare systems. Antiviral monoclonal antibodies (mAbs) are promising, high-added-value biotherapeutics. During recent years, the number of antiviral mAbs developed against both acute and chronic viruses has grown exponentially, some of them being currently tested in clinical trials. Antiviral mAbs can be used to blunt viral propagation through direct effects. They can also engage the host's immune system, leading to the induction of long-lasting protective vaccine-like effects. The assessment of mechanisms at play in the induction of vaccine-like effects by antiviral mAbs will help in improving antiviral treatments. Exploiting this effect will translate into therapeutic benefit for patients. The benefit will also help healthcare systems through the reduction of treatment costs.
Collapse
Affiliation(s)
- Mireia Pelegrin
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France.
| | - Mar Naranjo-Gomez
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| | - Marc Piechaczyk
- Equipe Labellisée par la Ligue contre le Cancer - Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, Université de Montpellier, 163 rue Auguste Broussonnet, 34090 Montpellier, France
| |
Collapse
|
12
|
Nwosu C, Yau HK, Becht S. Assignment of Core versus Antenna Fucosylation Types in Protein N-Glycosylation via Procainamide Labeling and Tandem Mass Spectrometry. Anal Chem 2015; 87:5905-13. [DOI: 10.1021/ac5040743] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Charles Nwosu
- Pharmaceutical Product Development, 8551 Research Way, Middleton, Wisconsin 53562, United States
| | - Hoi Kei Yau
- Pharmaceutical Product Development, 8551 Research Way, Middleton, Wisconsin 53562, United States
| | - Steven Becht
- Pharmaceutical Product Development, 8551 Research Way, Middleton, Wisconsin 53562, United States
| |
Collapse
|
13
|
Gomathinayagam S, Laface D, Houston-Cummings NR, Mangadu R, Moore R, Shandil I, Sharkey N, Li H, Stadheim TA, Zha D. In vivo anti-tumor efficacy of afucosylated anti-CS1 monoclonal antibody produced in glycoengineered Pichia pastoris. J Biotechnol 2015; 208:13-21. [PMID: 26015261 DOI: 10.1016/j.jbiotec.2015.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/21/2015] [Accepted: 05/13/2015] [Indexed: 11/15/2022]
Abstract
Monoclonal antibody (mAb) therapy has been successfully used for the treatment of B-cell lymphomas and is currently extended for the treatment of multiple myeloma (MM). New developments in MM therapeutics have achieved significant survival gains in patients but the disease still remains incurable. Elotuzumab (HuLuc63), an anti-CS1 monoclonal IgG1 antibody, is believed to induce anti-tumor activity and MM cytotoxicity through antibody dependent cellular cytotoxicity (ADCC) and inhibition of MM cell adhesion to bone marrow stromal cells (BMSCs). Modulations of the Fc glycan composition at the N297 site by selective mutations or afucosylation have been explored as strategies to develop bio-better therapeutics with enhanced ADCC activity. Afucosylated therapeutic antibodies with enhanced ADCC activity have been reported to possess greater efficacy in tumor growth inhibition at lower doses when compared to fucosylated therapeutic antibodies. The N-linked glycosylation pathway in Pichia pastoris has been engineered to produce human-like N-linked glycosylation with uniform afucosylated complex type glycans. The purpose of this study was to compare afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris with fucosylated anti-CS1 mAb expressed in mammalian HEK293 cells through in vitro ADCC and in vivo tumor inhibition models. Our results indicate that Fc glycosylation is critical for in vivo efficacy and afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris shows a better in vivo efficacy in tumor regression when compared to fucosylated anti-CS1 mAb expressed in HEK293 cells. Glycoengineered Pichia pastoris could provide an alternative platform for generating homogeneous afucosylated recombinant antibodies where Fc mediated immune effector function is important for efficacy.
Collapse
Affiliation(s)
- Sujatha Gomathinayagam
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Drake Laface
- Biologics Discovery, Palo Alto, Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, United States
| | - Nga Rewa Houston-Cummings
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Ruban Mangadu
- Biologics Discovery, Palo Alto, Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, United States
| | - Renee Moore
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Ishaan Shandil
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Nathan Sharkey
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Huijuan Li
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Terrance A Stadheim
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States
| | - Dongxing Zha
- GlycoFi Inc., A Wholly-Owned Subsidiary of Merck & Co Inc., 16 Cavendish Court, Lebanon, NH 03766, United States.
| |
Collapse
|
14
|
|
15
|
The genomic sequence of the Chinese hamster ovary (CHO)-K1 cell line. Nat Biotechnol 2011; 29:735-41. [PMID: 21804562 PMCID: PMC3164356 DOI: 10.1038/nbt.1932] [Citation(s) in RCA: 588] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 07/05/2011] [Indexed: 11/25/2022]
Abstract
Since 1987, immortalized cells from the ovary of a Chinese hamster have been the workhorse for producing recombinant therapeutics, including monoclonal antibodies, blood factors, hormones, growth factors and enzymes. Xu et al. provide the genome sequence of the ancestral CHO-K1 cell line, which should aid in the optimization of current production cell lines. Chinese hamster ovary (CHO)–derived cell lines are the preferred host cells for the production of therapeutic proteins. Here we present a draft genomic sequence of the CHO-K1 ancestral cell line. The assembly comprises 2.45 Gb of genomic sequence, with 24,383 predicted genes. We associate most of the assembled scaffolds with 21 chromosomes isolated by microfluidics to identify chromosomal locations of genes. Furthermore, we investigate genes involved in glycosylation, which affect therapeutic protein quality, and viral susceptibility genes, which are relevant to cell engineering and regulatory concerns. Homologs of most human glycosylation-associated genes are present in the CHO-K1 genome, although 141 of these homologs are not expressed under exponential growth conditions. Many important viral entry genes are also present in the genome but not expressed, which may explain the unusual viral resistance property of CHO cell lines. We discuss how the availability of this genome sequence may facilitate genome-scale science for the optimization of biopharmaceutical protein production.
Collapse
|
16
|
Zhang N, Liu L, Dumitru CD, Cummings NRH, Cukan M, Jiang Y, Li Y, Li F, Mitchell T, Mallem MR, Ou Y, Patel RN, Vo K, Wang H, Burnina I, Choi BK, Huber H, Stadheim TA, Zha D. Glycoengineered Pichia produced anti-HER2 is comparable to trastuzumab in preclinical study. MAbs 2011; 3:289-98. [PMID: 21487242 PMCID: PMC3149709 DOI: 10.4161/mabs.3.3.15532] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 03/19/2011] [Indexed: 01/12/2023] Open
Abstract
Mammalian cell culture systems are used predominantly for the production of therapeutic monoclonal antibody (mAb) products. A number of alternative platforms, such as Pichia engineered with a humanized N-linked glycosylation pathway, have recently been developed for the production of mAbs. The glycosylation profiles of mAbs produced in glycoengineered Pichia are similar to those of mAbs produced in mammalian systems. This report presents for the first time the comprehensive characterization of an anti-human epidermal growth factor receptor 2 (HER2) mAb produced in a glycoengineered Pichia, and a study comparing the anti-HER2 from Pichia, which had an amino acid sequence identical to trastuzumab, with trastuzumab. The comparative study covered a full spectrum of preclinical evaluation, including bioanalytical characterization, in vitro biological functions, in vivo anti-tumor efficacy and pharmacokinetics in both mice and non-human primates. Cell signaling and proliferation assays showed that anti-HER2 from Pichia had antagonist activities comparable to trastuzumab. However, Pichia-produced material showed a 5-fold increase in binding affinity to FcγIIIA and significantly enhanced antibody dependant cell-mediated cytotoxicity (ADCC) activity, presumably due to the lack of fucose on N-glycans. In a breast cancer xenograft mouse model, anti-HER2 was comparable to trastuzumab in tumor growth inhibition. Furthermore, comparable pharmacokinetic profiles were observed for anti-HER2 and trastuzumab in both mice and cynomolgus monkeys. We conclude that glycoengineered Pichia provides an alternative production platform for therapeutic mAbs and may be of particular interest for production of antibodies for which ADCC is part of the clinical mechanism of action.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized/immunology
- Antibody Affinity/immunology
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antibody-Dependent Cell Cytotoxicity/immunology
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- Area Under Curve
- Binding, Competitive/immunology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cells, Cultured
- Drug Evaluation, Preclinical
- Fucose/metabolism
- Genetic Engineering
- Humans
- Macaca fascicularis
- Mice
- Mice, Inbred C57BL
- Pichia/genetics
- Pichia/metabolism
- Polysaccharides/metabolism
- Protein Binding/immunology
- Receptor, ErbB-2/immunology
- Receptors, IgG/immunology
- Receptors, IgG/metabolism
- Recombinant Proteins/immunology
- Recombinant Proteins/pharmacokinetics
- Recombinant Proteins/pharmacology
- Trastuzumab
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ningyan Zhang
- Department of Biologics Research; Merck Research Laboratories; West Point, PA USA
| | - Liming Liu
- Drug Metabolism; Merck Research Laboratories; West Point, PA USA
| | - Calin Dan Dumitru
- Department of Biologics Research; Merck Research Laboratories; West Point, PA USA
| | | | | | | | - Yuan Li
- Department of Biologics Research; Merck Research Laboratories; West Point, PA USA
| | - Fang Li
- GlycoFi Inc.; Lebanon, NH USA
| | | | | | - Yangsi Ou
- Department of Biologics Research; Merck Research Laboratories; West Point, PA USA
| | | | - Kim Vo
- Department of Biologics Research; Merck Research Laboratories; West Point, PA USA
| | - Hui Wang
- Department of Biologics Research; Merck Research Laboratories; West Point, PA USA
| | | | | | - Hans Huber
- Department of Biologics Research; Merck Research Laboratories; West Point, PA USA
| | | | - Dongxing Zha
- Drug Metabolism; Merck Research Laboratories; West Point, PA USA
| |
Collapse
|
17
|
Jefferis R. The antibody paradigm: present and future development as a scaffold for biopharmaceutical drugs. Biotechnol Genet Eng Rev 2011; 26:1-42. [PMID: 21415874 DOI: 10.5661/bger-26-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Early studies of the humoral immune response revealed an apparent paradox: an infinite diversity of antibody specificities encoded within a finite genome. In consequence antibodies became a focus of interest for biochemists and geneticists. It resulted in the elucidation of the basic structural unit, the immunoglobulin (Ig) domain, comprised of ~ 100 amino acid residues that generate the characteristic "immunoglobulin (Ig) fold". The Ig fold has an anti-parallel ß-pleated sheet (barrel) structure that affords structural stability whilst the ß-bends allow for essentially infinite structural variation and functional diversity. This versatility is reflected in the Ig domain being the most widely utilised structural unit within the proteome. Human antibodies are comprised of multiple Ig domains and their structural diversity may be enhanced through the attachment of oligosaccharides. This review summarizes our current understanding of the immunoglobulin structure/function relationships and the application of protein and oligosaccharide engineering to further develop the Ig domain as a scaffold for the generation of new and novel antibody based therapeutics.
Collapse
Affiliation(s)
- Roy Jefferis
- School of Immunity and Infection, The College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK.
| |
Collapse
|
18
|
|
19
|
Abstract
Bispecific antibodies, in contrast to conventional monoclonal antibodies, can bind simultaneously two different antigens. Taking advantage of this virtue, they are mostly designed for immune effector cell redirection to tumors and for radionuclide pretargeting to tumors. Bispecific antibodies of the first generation were produced by chemical cross-linking or cell-fusion technologies. More recently, the application of genetic engineering technologies gave rise to numerous formats of bispecific antibody fragments and whole IgG molecules. Because bispecific antibodies enable therapeutic strategies that are not possible with conventional monoclonal antibodies, they attract strong interest. Several bispecific antibody formats have already shown clinical efficacy in cancer patients, catalyzing efforts to translate the imaginative bispecific antibody concepts into effective therapies.
Collapse
Affiliation(s)
- Nurit Hollander
- Department of Human Microbiology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
20
|
Natsume A, Shimizu-Yokoyama Y, Satoh M, Shitara K, Niwa R. Engineered anti-CD20 antibodies with enhanced complement-activating capacity mediate potent anti-lymphoma activity. Cancer Sci 2009; 100:2411-8. [PMID: 19758394 PMCID: PMC11159923 DOI: 10.1111/j.1349-7006.2009.01327.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
One of the major issues in current antibody therapy is insufficient efficacy. Various biological factors relating to the host's immune system or tumor cells have been suggested to reduce the efficacy of anti-CD20 therapy in B-cell malignancies. In this study, we characterized the in vitro anti-lymphoma activity of anti-CD20 antibodies having a novel engineered heavy chain with enhanced complement-dependent cytotoxicity (CDC). Anti-CD20 antibodies having a variant heavy constant region of mixed IgG1/IgG3 isotype, which have previously been found to enhance CDC, were investigated for their in vitro CDC against lymphoma cells and whole blood B-cell depletion activity. Use of the variant constant region greatly increased the CDC of an anti-CD20 antibody having variable regions identical to those of rituximab to the level shown by an IgG1 antibody of ofatumumab. Although the whole blood assay showed different cytotoxicity patterns among individual blood donors, the CDC-enhancing variant of rituximab showed higher activity than the parent IgG1 and consistently showed maximized activity when further combined with antibody-dependent cellular cytotoxicity (ADCC)-enhancing modification by fucose removal from Fc-linked oligosaccharides. In addition, the rituximab variant showed potent CDC against transfectant cells with lower CD20 expression and chronic lymphocytic leukemia-derived cell lines with higher complement regulatory proteins. These findings suggest that CDC enhancement, both alone and in combination with ADCC enhancement, increases the anti-lymphoma activity of anti-CD20 antibodies irrespective of individual differences in effector functions, and renders current anti-CD20 therapy capable of overcoming the potential resistance mechanisms.
Collapse
Affiliation(s)
- Akito Natsume
- Antibody Research Laboratories, Research Division, Kyowa Hakko Kirin Co, Ltd, Machida-shi, Tokyo, Japan
| | | | | | | | | |
Collapse
|
21
|
Esue O, Kanai S, Liu J, Patapoff TW, Shire SJ. Carboxylate-Dependent Gelation of a Monoclonal Antibody. Pharm Res 2009; 26:2478-85. [DOI: 10.1007/s11095-009-9963-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
22
|
Yamane-Ohnuki N, Satoh M. Production of therapeutic antibodies with controlled fucosylation. MAbs 2009; 1:230-6. [PMID: 20065644 DOI: 10.4161/mabs.1.3.8328] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The clinical success of therapeutic antibodies is demonstrated by the number of antibody therapeutics that have been brought to market and the increasing number of therapeutic antibodies in development. Recombinant antibodies are molecular-targeted therapeutic agents and represent a major new class of drugs. However, it is still very important to optimize and maximize the clinical efficacy of therapeutic antibodies, in part to help lower the cost of therapeutic antibodies by potentially reducing the dose or the duration of treatment. Clinical trials using therapeutic antibodies fully lacking core fucose residue in the Fc oligosaccharides are currently underway, and their remarkable physiological activities in humans in vivo have attracted attention as next-generation therapeutic antibody approaches with improved efficacy. Thus, an industrially applicable antibody production process that provides consistent yields of fully non-fucosylated antibody therapeutics with fixed quality has become a key goal in the successful development of next-generation therapeutic agents. In this article, we review the current technologies for production of therapeutic antibodies with control of fucosylation of the Fc N-glycans.
Collapse
Affiliation(s)
- Naoko Yamane-Ohnuki
- Antibody Research Laboratories, Research Division, Kyowa Hakko Kirin Co., Ltd., Machida-shi, Tokyo, Japan
| | | |
Collapse
|
23
|
Weisser NE, Hall JC. Applications of single-chain variable fragment antibodies in therapeutics and diagnostics. Biotechnol Adv 2009; 27:502-20. [PMID: 19374944 DOI: 10.1016/j.biotechadv.2009.04.004] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 03/03/2009] [Accepted: 04/04/2009] [Indexed: 11/18/2022]
Abstract
Antibodies (Abs) are some of the most powerful tools in therapy and diagnostics and are currently one of the fastest growing classes of therapeutic molecules. Recombinant antibody (rAb) fragments are becoming popular therapeutic alternatives to full length monoclonal Abs since they are smaller, possess different properties that are advantageous in certain medical applications, can be produced more economically and are easily amendable to genetic manipulation. Single-chain variable fragment (scFv) Abs are one of the most popular rAb format as they have been engineered into larger, multivalent, bi-specific and conjugated forms for many clinical applications. This review will show the tremendous versatility and importance of scFv fragments as they provide the basic antigen binding unit for a multitude of engineered Abs for use as human therapeutics and diagnostics.
Collapse
Affiliation(s)
- Nina E Weisser
- Department of Environmental Biology, University of Guelph, Guelph, ON N1G2W1, Canada
| | | |
Collapse
|
24
|
|
25
|
Jain M, Kamal N, Batra SK. Engineering antibodies for clinical applications. Trends Biotechnol 2007; 25:307-16. [PMID: 17512622 DOI: 10.1016/j.tibtech.2007.05.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Revised: 03/07/2007] [Accepted: 05/04/2007] [Indexed: 11/17/2022]
Abstract
Molecular engineering has contributed immensely to the clinical success of antibodies in recent years. The modular structure of antibodies has permitted their modification in numerous ways, to meet various clinical requirements. With the help of antibody engineering, it has been possible to modify the molecular size, pharmacokinetics, immunogenicity, binding affinity, specificity and effector function of antibodies. In addition, fusion proteins of antibodies with various proteins and peptides have yielded targeted biological modifiers, toxins and imaging agents. This review focuses on the recent trends in antibody engineering for improving their clinical utility.
Collapse
Affiliation(s)
- Maneesh Jain
- Department of Biochemistry and Molecular Biology, Department of Pathology and Microbiology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | | | | |
Collapse
|
26
|
Kuystermans D, Krampe B, Swiderek H, Al-Rubeai M. Using cell engineering and omic tools for the improvement of cell culture processes. Cytotechnology 2007; 53:3-22. [PMID: 19003186 DOI: 10.1007/s10616-007-9055-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 01/25/2007] [Indexed: 12/26/2022] Open
Abstract
Significant strides have been made in mammalian cell based biopharmaceutical process and cell line development over the past years. With several established mammalian host cell lines and expression systems, optimization of selection systems to reduce development times and improvement of glycosylation patterns are only some of the advances being made to improve cell culture processes. In this article, the advances pertaining to cell line development and cell engineering strategies are discussed. An overview of the cell engineering strategies to enhance cellular characteristics by genetic manipulation are illustrated, focusing on the use of genomics and proteomics tools and their application in such endeavors. Included in this review are some of the early studies using the 'omic' technique to understand cellular mechanisms of product synthesis and secretion, apoptosis, cell proliferation and the influence of the physicochemical environment. The article highlights the significance of integrating genomics and proteomics data with the vast amounts of bioprocess data for improved analysis of the biological pathways involved. Further improvements of the techniques and methodologies used are needed but ultimately, the new cell engineering strategies should provide great insight into the regulatory networks within the cell in a bioprocess environment and how to manipulate them to increase overall productivity.
Collapse
Affiliation(s)
- Darrin Kuystermans
- School of Chemical and Bioprocess Engineering and Centre for Synthesis and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | |
Collapse
|