1
|
Page A, Fusil F, Cosset FL. Towards Physiologically and Tightly Regulated Vectored Antibody Therapies. Cancers (Basel) 2020; 12:E962. [PMID: 32295072 PMCID: PMC7226531 DOI: 10.3390/cancers12040962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/20/2022] Open
Abstract
Cancers represent highly significant health issues and the options for their treatment are often not efficient to cure the disease. Immunotherapy strategies have been developed to modulate the patient's immune system in order to eradicate cancerous cells. For instance, passive immunization consists in the administration at high doses of exogenously produced monoclonal antibodies directed either against tumor antigen or against immune checkpoint inhibitors. Its main advantage is that it provides immediate immunity, though during a relatively short period, which consequently requires frequent injections. To circumvent this limitation, several approaches, reviewed here, have emerged to induce in vivo antibody secretion at physiological doses. Gene delivery vectors, such as adenoviral vectors or adeno-associated vectors, have been designed to induce antibody secretion in vivo after in situ cell modification, and have driven significant improvements in several cancer models. However, anti-idiotypic antibodies and escape mutants have been detected, probably because of both the continuous expression of antibodies and their expression by unspecialized cell types. To overcome these hurdles, adoptive transfer of genetically modified B cells that secrete antibodies either constitutively or in a regulated manner have been developed by ex vivo transgene insertion with viral vectors. Recently, with the emergence of gene editing technologies, the endogenous B cell receptor loci of B cells have been modified with the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated endonuclease (Cas-9) system to change their specificity in order to target a given antigen. The expression of the modified BCR gene hence follows the endogenous regulation mechanisms, which may prevent or at least reduce side effects. Although these approaches seem promising for cancer treatments, major questions, such as the persistence and the re-activation potential of these engineered cells, remain to be addressed in clinically relevant animal models before translation to humans.
Collapse
Affiliation(s)
| | | | - François-Loïc Cosset
- CIRICentre International de Recherche en Infectiologie, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46 allée d’Italie, F-69007 Lyon, France; (A.P.); (F.F.)
| |
Collapse
|
2
|
Matsunaga S, Jeremiah SS, Miyakawa K, Kurotaki D, Shizukuishi S, Watashi K, Nishitsuji H, Kimura H, Tamura T, Yamamoto N, Shimotohno K, Wakita T, Ryo A. Engineering Cellular Biosensors with Customizable Antiviral Responses Targeting Hepatitis B Virus. iScience 2020; 23:100867. [PMID: 32105634 PMCID: PMC7113479 DOI: 10.1016/j.isci.2020.100867] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/16/2019] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
SynNotch receptor technology is a versatile tool that uses the regulatory notch core portion with an extracellular scFv and an intracellular transcription factor that enables to program customized input and output functions in mammalian cells. In this study, we designed a novel synNotch receptor comprising scFv against HBs antigen linked with an intracellular artificial transcription factor and exploited it for viral sensing and cellular immunotherapy. The synNotch receptor expressing cells sensed HBV particles and membrane-bound HBs antigens and responded by expressing reporter molecules, secNL or GFP. We also programmed these cells to dispense antiviral responses such as type I interferon and anti-HBV neutralizing mouse-human chimeric antibodies. Our data reveal that synNotch receptor signaling works for membrane-bound ligands such as enveloped viral particles and proteins borne on liposomal vesicles. This study establishes the concepts of "engineered immunity" where the synNotch platform is utilized for cellular immunotherapy against viral infections.
Collapse
Affiliation(s)
- Satoko Matsunaga
- Department of Microbiology, Yokohama City University School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Sundararaj S Jeremiah
- Department of Microbiology, Yokohama City University School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Kei Miyakawa
- Department of Microbiology, Yokohama City University School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Daisuke Kurotaki
- Department of Immunology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Sayaka Shizukuishi
- Department of Microbiology, Yokohama City University School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Hironori Nishitsuji
- Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba 272-8516, Japan
| | - Hirokazu Kimura
- School of Medical Technology, Faculty of Health Sciences, Gunma Paz University, Takasaki 370-0006, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Naoki Yamamoto
- Department of Microbiology, Yokohama City University School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Kunitada Shimotohno
- Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba 272-8516, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan.
| |
Collapse
|
3
|
Production and Application of Multicistronic Constructs for Various Human Disease Therapies. Pharmaceutics 2019; 11:pharmaceutics11110580. [PMID: 31698727 PMCID: PMC6920891 DOI: 10.3390/pharmaceutics11110580] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/30/2019] [Accepted: 11/03/2019] [Indexed: 01/09/2023] Open
Abstract
The development of multicistronic vectors has opened up new opportunities to address the fundamental issues of molecular and cellular biology related to the need for the simultaneous delivery and joint expression of several genes. To date, the examples of the successful use of multicistronic vectors have been described for the development of new methods of treatment of various human diseases, including cardiovascular, oncological, metabolic, autoimmune, and neurodegenerative disorders. The safety and effectiveness of the joint delivery of therapeutic genes in multicistronic vectors based on the internal ribosome entry site (IRES) and self-cleaving 2A peptides have been shown in both in vitro and in vivo experiments as well as in clinical trials. Co-expression of several genes in one vector has also been used to create animal models of various inherited diseases which are caused by mutations in several genes. Multicistronic vectors provide expression of all mutant genes, which allows the most complete mimicking disease pathogenesis. This review comprehensively discusses multicistronic vectors based on IRES nucleotide sequence and self-cleaving 2A peptides, including its features and possible application for the treatment and modeling of various human diseases.
Collapse
|
4
|
Fallah A, Estiri H, Parrish E, Soleimani M, Zeinali S, Zadeh-Vakili A. Biosimilar Gene Therapy: Investigational Assessment of Secukinumab Gene Therapy. CELL JOURNAL 2019; 21:433-443. [PMID: 31376325 PMCID: PMC6722441 DOI: 10.22074/cellj.2020.6309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 12/09/2018] [Indexed: 11/20/2022]
Abstract
Objective Tumor necrosis factor-alpha (TNF-α), checkpoint inhibitors, and interleukin-17 (IL-17) are critical targets in
inflammation and autoimmune diseases. Monoclonal antibodies (mAbs) have a successful portfolio in the treatment of chronic
diseases. With the current progress in stem cells and gene therapy technologies, there is the promise of replacing costly mAbs
production in bioreactors with a more direct and cost-effective production method inside the patient’s cells. In this paper we
examine the results of an investigational assessment of secukinumab gene therapy.
Materials and Methods In this experimental study, the DNA sequence of the heavy and light chains of secukinumab
antibodies were cloned in a lentiviral vector. Human chorionic villous mesenchymal stem cells (CMSCs) were isolated and
characterized. After lentiviral packaging and titration, part of the recombinant viruses was used for transduction of the CMSCs
and the other part were applied for systemic gene therapy. The engineered stem cells and recombinant viruses were applied
for ex vivo and in vivo gene therapy, respectively, in different groups of rat models. In vitro and in vivo secukinumab expression
was confirmed with quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and ELISA by considering the
approved secukinumab as the standard reference.
Results Cell differentiation assays and flow cytometry of standard biomarkers confirmed the multipotency of the
CMSCs. Western blot and qRT-PCR confirmed in vitro gene expression of secukinumab at both the mRNA and protein
level. ELISA testing of serum from treated rat models confirmed mAb overexpression for both in vivo and ex vivo gene
therapies.
Conclusion In this study, a lentiviral-mediated ex vivo and in vivo gene therapy was developed to provide a moderate dose
of secukinumab in rat models. Biosimilar gene therapy is an attractive approach for the treatment of autoimmune disorders,
cancers and other chronic diseases.
Collapse
Affiliation(s)
- Ali Fallah
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,RNAx Ltd., London, UK
| | | | | | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sirous Zeinali
- Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran.Electronic Address:
| | - Azita Zadeh-Vakili
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.Electronic Address:
| |
Collapse
|
5
|
Zhang S, Zheng C, Zhu W, Xiong P, Zhou D, Huang C, Zheng D. A novel anti-DR5 antibody-drug conjugate possesses a high-potential therapeutic efficacy for leukemia and solid tumors. Am J Cancer Res 2019; 9:5412-5423. [PMID: 31410224 PMCID: PMC6691585 DOI: 10.7150/thno.33598] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/21/2019] [Indexed: 12/17/2022] Open
Abstract
It is well known that tumor necrosis factor-related apoptosis inducing ligand receptor 1 or 2 (DR4/DR5) is specifically expressed in various tumor cells, but less or no expression in most normal cells. Many first generations of TRAIL agonists including recombinant preparations of TRAIL, agonistic antibodies against DR4/DR5 have been developed in phase I/II clinical trials for cancer therapy. However, the outcomes of clinical trials by using DR4/DR5 agonist mono-therapy were disappointed even though the safety profile was well tolerance. In the present study, we report an anti-DR5 antibody-drug conjugate (ADC, named as Zapadcine-1) possesses a higher potential for the therapy of lymphocyte leukemia and solid cancers. Methods: Zapadcine-1 was made by a fully humanized DR5-specific monoclonal antibody (Zaptuzumab) coupled via a cleavable linker to a highly toxic inhibitor of tubulin, monomethyl auristatin D (MMAD), by using ThioBridge technology. Cytotoxicity of the ADC in various tumor cells was identified by luminescent cell viability assay and the efficacy in vivo was determined in cells derived xenografts (CDX) of Jurkat E6-1, BALL-1, Reh, and patient derived xenografts (PDX) of human acute leukemia. Preliminary safety evaluation was carried out in rat and monkey. Results: Zapadcine-1 possesses a similar binding ability to the death receptor DR5 as the naked monoclonal antibody Zaptuzumab, and can be rapidly endocytosed into the lysosome of cancer cells. Zapadcine-1 specifically kills human lymphocyte leukemia cells and solid tumor cells, but not normal cells tested. More importantly, Zapadcine-1 drastically eliminates the xenografts in both CDX and PDX models of human acute leukemia. The excellent and comparable therapeutic efficacy is also observed in lung cancer NCI-H1975 CDX mouse model. The maximum-tolerated dose (MTD) of single injected Zapadcine-1 in rat and cynomolgus monkey shows an acceptable safety profile. Conclusion: These data demonstrate a promising anti-cancer activity, meriting further exploration of its potential as a novel cancer therapeutic agent, especially for the acute lymphocyte leukemia.
Collapse
|
6
|
Moradi Marjaneh R, Hassanian SM, Ghobadi N, Ferns GA, Karimi A, Jazayeri MH, Nasiri M, Avan A, Khazaei M. Targeting the death receptor signaling pathway as a potential therapeutic target in the treatment of colorectal cancer. J Cell Physiol 2018; 233:6538-6549. [DOI: 10.1002/jcp.26640] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/30/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Reyhaneh Moradi Marjaneh
- Department of Physiology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Medical Biochemistry, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Microanatomy Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Niloofar Ghobadi
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School Division of Medical Education Falmer, Brighton, Sussex UK
| | - Afshin Karimi
- Quality Department of Nutricia Mashhad Mild Powder Industrial Mashhad Iran
| | - Mir Hadi Jazayeri
- Immunology Research Center and Department of Immunology, School of Medicine Iran University of Medical Sciences Tehran Iran
| | - Mohammadreza Nasiri
- Recombinant Proteins Research Group The Research Institute of Biotechnology, Ferdowsi University of Mashhad Mashhad Iran
| | - Amir Avan
- Metabolic Syndrome Research Center Mashhad University of Medical Sciences Mashhad Iran
- Cancer Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Modern Sciences and Technologies, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
- Surgical Oncology Research Center Mashhad University of Medical Sciences Mashhad Iran
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
7
|
Ebadat S, Ahmadi S, Ahmadi M, Nematpour F, Barkhordari F, Mahdian R, Davami F, Mahboudi F. Evaluating the efficiency of CHEF and CMV promoter with IRES and Furin/2A linker sequences for monoclonal antibody expression in CHO cells. PLoS One 2017; 12:e0185967. [PMID: 29023479 PMCID: PMC5638317 DOI: 10.1371/journal.pone.0185967] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 09/24/2017] [Indexed: 11/19/2022] Open
Abstract
In recent years, monoclonal antibodies (mAbs) have been developed as powerful therapeutic and diagnostic agents and Chinese hamster ovary (CHO) cells have emerged as the dominant host for the recombinant expression of these proteins. A critical step in recombinant expression is the utilization of strong promoters, such as the Chinese Hamster Elongation Factor-1α (CHEF-1) promoter. To compare the strengths of CHEF with cytomegalovirus (CMV) promoter for mAb expression in CHO cells, four bicistronic vectors bearing either internal ribosome entry site (IRES) or Furin/2A (F2A) sequences were designed. The efficiency of these promoters was evaluated by measuring level of expressed antibody in stable cell pools. Our results indicated that CHEF promoter-based expression of mAbs was 2.5 fold higher than CMV-based expression in F2A-mediated vectors. However, this difference was less significant in IRES-mediated mAb expressing cells. Studying the stability of the F2A expression system in the course of 18 weeks, we observed that the cells having CHEF promoter maintained their antibody expression at higher level than those transfected with CMV promoter. Further analyses showed that both IRES-mediated vectors, expressed mAbs with correct size, whereas in antibodies expressed via F2A system heterogeneity of light chains were detected due to incomplete furin cleavage. Our findings indicated that the CHEF promoter is a viable alternative to CMV promoter-based expression in F2A-mediated vectors by providing both higher expression and level of stability.
Collapse
Affiliation(s)
- Saeedeh Ebadat
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Samira Ahmadi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Ahmadi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Medical Biotechnology Department, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Nematpour
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Reza Mahdian
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
8
|
Lin J, Neo SH, Ho SCL, Yeo JHM, Wang T, Zhang W, Bi X, Chao SH, Yang Y. Impact of Signal Peptides on Furin-2A Mediated Monoclonal Antibody Secretion in CHO Cells. Biotechnol J 2017; 12. [DOI: 10.1002/biot.201700268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/03/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Jian'er Lin
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
| | - Shu Hui Neo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
| | - Steven C. L. Ho
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
| | - Jessna H. M. Yeo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
| | - Tianhua Wang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
| | - Wei Zhang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
| | - Xuezhi Bi
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
| | - Sheng-Hao Chao
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
- Department of Microbiology; National University of Singapore; Block MD4, 5 Science Drive 2 Singapore 117597 Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR); 20 Biopolis Way, #06-01 Centros Singapore 138668 Singapore
| |
Collapse
|
9
|
Ahmadi S, Davami F, Davoudi N, Nematpour F, Ahmadi M, Ebadat S, Azadmanesh K, Barkhordari F, Mahboudi F. Monoclonal antibodies expression improvement in CHO cells by PiggyBac transposition regarding vectors ratios and design. PLoS One 2017; 12:e0179902. [PMID: 28662065 PMCID: PMC5491063 DOI: 10.1371/journal.pone.0179902] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/06/2017] [Indexed: 12/22/2022] Open
Abstract
Establishing stable Chinese Hamster Ovary (CHO) cells producing monoclonal antibodies (mAbs) usually pass through the random integration of vectors to the cell genome, which is sensitive to gene silencing. One approach to overcome this issue is to target a highly transcribed region in the genome. Transposons are useful devices to target active parts of genomes, and PiggyBac (PB) transposon can be considered as a good option. In the present study, three PB transposon donor vectors containing both heavy and light chains were constructed, one contained independent expression cassettes while the others utilized either an Internal Ribosome Entry Site (IRES) or 2A element to express mAb. Conventional cell pools were created by transferring donor vectors into the CHO cells, whereas transposon-based cells were generated by transfecting the cells with donor vectors with a companion of a transposase-encoding helper vector, with 1:2.5 helper/donor vectors ratio. To evaluate the influence of helper/donor vectors ratio on expression, the second transposon-based cell pools were generated with 1:5 helper/donor ratio. Expression levels in the transposon-based cells were two to five -folds more than those created by conventional method except for the IRES-mediated ones, in which the observed difference increased more than 100-fold. The results were dependent on both donor vector design and vectors ratios.
Collapse
Affiliation(s)
- Samira Ahmadi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Davami
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Noushin Davoudi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Nematpour
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Maryam Ahmadi
- Medical Biotechnology Department, Semnan University of Medical Sciences, Semnan, Iran
| | - Saeedeh Ebadat
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Fereidoun Mahboudi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- * E-mail: ,
| |
Collapse
|
10
|
Hollevoet K, Declerck PJ. State of play and clinical prospects of antibody gene transfer. J Transl Med 2017; 15:131. [PMID: 28592330 PMCID: PMC5463339 DOI: 10.1186/s12967-017-1234-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/31/2017] [Indexed: 12/31/2022] Open
Abstract
Recombinant monoclonal antibodies (mAbs) are one of today's most successful therapeutic classes in inflammatory diseases and oncology. A wider accessibility and implementation, however, is hampered by the high product cost and prolonged need for frequent administration. The surge in more effective mAb combination therapies further adds to the costs and risk of toxicity. To address these issues, antibody gene transfer seeks to administer to patients the mAb-encoding nucleotide sequence, rather than the mAb protein. This allows the body to produce its own medicine in a cost- and labor-effective manner, for a prolonged period of time. Expressed mAbs can be secreted systemically or locally, depending on the production site. The current review outlines the state of play and clinical prospects of antibody gene transfer, thereby highlighting recent innovations, opportunities and remaining hurdles. Different expression platforms and a multitude of administration sites have been pursued. Viral vector-mediated mAb expression thereby made the most significant strides. Therapeutic proof of concept has been demonstrated in mice and non-human primates, and intramuscular vectored mAb therapy is under clinical evaluation. However, viral vectors face limitations, particularly in terms of immunogenicity. In recent years, naked DNA has gained ground as an alternative. Attained serum mAb titers in mice, however, remain far below those obtained with viral vectors, and robust pharmacokinetic data in larger animals is limited. The broad translatability of DNA-based antibody therapy remains uncertain, despite ongoing evaluation in patients. RNA presents another emerging platform for antibody gene transfer. Early reports in mice show that mRNA may be able to rival with viral vectors in terms of generated serum mAb titers, although expression appears more short-lived. Overall, substantial progress has been made in the clinical translation of antibody gene transfer. While challenges persist, clinical prospects are amplified by ongoing innovations and the versatility of antibody gene transfer. Clinical introduction can be expedited by selecting the platform approach currently best suited for the mAb or disease of interest. Innovations in expression platform, administration and antibody technology are expected to further improve overall safety and efficacy, and unlock the vast clinical potential of antibody gene transfer.
Collapse
Affiliation(s)
- Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Campus Gasthuisberg O&N 2, P.B. 820, Herestraat 49, 3000 Leuven, Belgium
| | - Paul J. Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Campus Gasthuisberg O&N 2, P.B. 820, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
11
|
RANJBAR S, HASHEMZADEH MS, KHOSHTINAT NIKKHOI S, FARASAT A, TAT M, GHALAVAND M, DOROSTKAR R. Selective suppression of tumor cells by a tumor-specific bicistronic lentiviral vector. Turk J Biol 2016. [DOI: 10.3906/biy-1512-53] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
12
|
Chng J, Wang T, Nian R, Lau A, Hoi KM, Ho SCL, Gagnon P, Bi X, Yang Y. Cleavage efficient 2A peptides for high level monoclonal antibody expression in CHO cells. MAbs 2015; 7:403-12. [PMID: 25621616 DOI: 10.1080/19420862.2015.1008351] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Linking the heavy chain (HC) and light chain (LC) genes required for monoclonal antibodies (mAb) production on a single cassette using 2A peptides allows control of LC and HC ratio and reduces non-expressing cells. Four 2A peptides derived from the foot-and-mouth disease virus (F2A), equine rhinitis A virus (E2A), porcine teschovirus-1 (P2A) and Thosea asigna virus (T2A), respectively, were compared for expression of 3 biosimilar IgG1 mAbs in Chinese hamster ovary (CHO) cell lines. HC and LC were linked by different 2A peptides both in the absence and presence of GSG linkers. Insertion of a furin recognition site upstream of 2A allowed removal of 2A residues that would otherwise be attached to the HC. Different 2A peptides exhibited different cleavage efficiencies that correlated to the mAb expression level. The relative cleavage efficiency of each 2A peptide remains similar for expression of different IgG1 mAbs in different CHO cells. While complete cleavage was not observed for any of the 2A peptides, GSG linkers did enhance the cleavage efficiency and thus the mAb expression level. T2A with the GSG linker (GT2A) exhibited the highest cleavage efficiency and mAb expression level. Stably amplified CHO DG44 pools generated using GT2A had titers 357, 416 and 600 mg/L for the 3 mAbs in shake flask batch cultures. Incomplete cleavage likely resulted in incorrectly processed mAb species and aggregates, which were removed with a chromatin-directed clarification method and protein A purification. The vector and methods presented provide an easy process beneficial for both mAb development and manufacturing.
Collapse
Key Words
- 2A peptide
- CHO
- CHO, Chinese hamster ovary
- E2A, 2A peptide derived from the equine rhinitis virus
- F2A, 2A peptide derived from the foot-and-mouth disease virus
- G, glycine
- GE2A, E2A with the GSG linker
- GF2A, F2A with the GSG linker
- GFP, green fluorescence protein
- GP2A, P2A with the GSG linker
- GSG linker
- GT2A, T2A with the GSG linker
- HC, heavy chain
- HT, hypoxanthine and thymine
- IRES, internal ribosome entry site
- IgG, immunoglobulin G
- K, lysine
- LC, light chain
- MS, mass spectrometry
- MTX, methotrexate
- P, proline
- P2A, 2A peptide derived from the porcine teschovirus-1
- PFM, protein-free medium
- PVDF, polyvinylidene difluoride
- SEC, size exclusion chromatography
- T2A, 2A peptide derived from the Thosea asigna virus
- cleavage efficiency
- furin
- mAb, monoclonal antibody
- monoclonal antibody
Collapse
Affiliation(s)
- Jake Chng
- a Bioprocessing Technology Institute; Agency for Science , Technology and Research (A*STAR) ; Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
The use of antibodies as a treatment for disease has it origins in experiments performed in the 1890s, and since these initial experiments, monoclonal antibodies (mAbs) have become one of the fastest growing therapeutic classes for the treatment of cancer, autoimmune disease, and infectious diseases. However, treatment with therapeutic mAbs often requires high doses given via long infusions or multiple injections, which, coupled with the prohibitively high cost associated with the production of clinical-grade proteins and the transient serum half-lives that necessitate multiple administrations to gain therapeutic benefits, makes large-scale treatment of patients, especially patients in the developing world, difficult. Due to their low-cost and rapid scalability, nucleic acid-based approaches to deliver antibody gene sequences for in situ mAb production have gained substantial traction. In this review, we discuss new approaches to produce therapeutic mAbs in situ to overcome the need for the passive infusion of purified protein.
Collapse
Affiliation(s)
- Todd J Suscovich
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | | |
Collapse
|
14
|
Koehler BC, Jäger D, Schulze-Bergkamen H. Targeting cell death signaling in colorectal cancer: Current strategies and future perspectives. World J Gastroenterol 2014; 20:1923-1934. [PMID: 24587670 PMCID: PMC3934462 DOI: 10.3748/wjg.v20.i8.1923] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/06/2013] [Accepted: 01/15/2014] [Indexed: 02/06/2023] Open
Abstract
The evasion from controlled cell death induction has been considered as one of the hallmarks of cancer cells. Defects in cell death signaling are a fundamental phenomenon in colorectal cancer. Nearly any non-invasive cancer treatment finally aims to induce cell death. However, apoptosis resistance is the major cause for insufficient therapeutic success and disease relapse in gastrointestinal oncology. Various compounds have been developed and evaluated with the aim to meet with this obstacle by triggering cell death in cancer cells. The aim of this review is to illustrate current approaches and future directions in targeting cell death signaling in colorectal cancer. The complex signaling network of apoptosis will be demonstrated and the “druggability” of targets will be identified. In detail, proteins regulating mitochondrial cell death in colorectal cancer, such as Bcl-2 and survivin, will be discussed with respect to potential therapeutic exploitation. Death receptor signaling and targeting in colorectal cancer will be outlined. Encouraging clinical trials including cell death based targeted therapies for colorectal cancer are under way and will be demonstrated. Our conceptual understanding of cell death in cancer is rapidly emerging and new types of controlled cellular death have been identified. To meet this progress in cell death research, the implication of autophagy and necroptosis for colorectal carcinogenesis and therapeutic approaches will also be depicted. The main focus of this topic highlight will be on the revelation of the complex cell death concepts in colorectal cancer and the bridging from basic research to clinical use.
Collapse
|
15
|
Yang L, Wang P. Passive immunization against HIV/AIDS by antibody gene transfer. Viruses 2014; 6:428-47. [PMID: 24473340 PMCID: PMC3939464 DOI: 10.3390/v6020428] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 01/06/2014] [Accepted: 01/10/2014] [Indexed: 12/12/2022] Open
Abstract
Despite tremendous efforts over the course of many years, the quest for an effective HIV vaccine by the classical method of active immunization remains largely elusive. However, two recent studies in mice and macaques have now demonstrated a new strategy designated as Vectored ImmunoProphylaxis (VIP), which involves passive immunization by viral vector-mediated delivery of genes encoding broadly neutralizing antibodies (bnAbs) for in vivo expression. Robust protection against virus infection was observed in preclinical settings when animals were given VIP to express monoclonal neutralizing antibodies. This unorthodox approach raises new promise for combating the ongoing global HIV pandemic. In this article, we survey the status of antibody gene transfer, review the revolutionary progress on isolation of extremely bnAbs, detail VIP experiments against HIV and its related virus conduced in humanized mice and macaque monkeys, and discuss the pros and cons of VIP and its opportunities and challenges towards clinical applications to control HIV/AIDS endemics.
Collapse
Affiliation(s)
- Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California at Los Angeles, Los Angeles, CA 90095, USA.
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
16
|
Comparison of internal ribosome entry site (IRES) and Furin-2A (F2A) for monoclonal antibody expression level and quality in CHO cells. PLoS One 2013; 8:e63247. [PMID: 23704898 PMCID: PMC3660568 DOI: 10.1371/journal.pone.0063247] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 04/01/2013] [Indexed: 12/16/2022] Open
Abstract
Four versions of tricistronic vectors expressing IgG1 light chain (LC), IgG1 heavy chain (HC), and dihydrofolate reductase (DHFR) in one transcript were designed to compare internal ribosome entry site (IRES) and furin-2A (F2A) for their influence on monoclonal antibody (mAb) expression level and quality in CHO DG44 cells. LC and HC genes are arranged as either the first or the second cistron. When using mAb quantification methods based on the detection antibodies against HC Fc region, F2A-mediated tricistronic vectors appeared to express mAb at higher levels than the IRES-mediated tricistronic vectors in both transient and stable transfections. Further analysis revealed that more than 40% of products detected in stably transfected pools generated using the two F2A-mediated tricistronic vectors were aggregates. LC and HC from the F2A stably transfected pools were not properly processed, giving rise to LC+F2A+HC or HC+F2A+LC fusion proteins, LC and HC polypeptides with F2A remnants, and incorrectly cleaved signal peptides. Both IRES-mediated tricistronic vectors express mAb with correct sizes and signal peptide cleavage. Arrangement of LC as the first cistron in the IRES-mediated tricistronic vectors exhibits increased mAb expression level, better growth, and minimized product aggregation, while arrangement of HC as first cistron results in low expression, slower growth, and high aggregation. The results obtained will be beneficial for designing vectors that enhance mAb expression level and quality in mammalian cells.
Collapse
|
17
|
Jia LT, Chen SY, Yang AG. Cancer gene therapy targeting cellular apoptosis machinery. Cancer Treat Rev 2012; 38:868-76. [PMID: 22800735 DOI: 10.1016/j.ctrv.2012.06.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/21/2012] [Accepted: 06/22/2012] [Indexed: 01/14/2023]
Abstract
The unraveling of cellular apoptosis machinery provides novel targets for cancer treatment, and gene therapy targeting this suicidal system has been corroborated to cause inflammation-free autonomous elimination of neoplastic cells. The apoptotic machinery can be targeted by introduction of a gene encoding an inducer, mediator or executioner of apoptotic cell death or by inhibition of anti-apoptotic gene expression. Strategies targeting cancer cells, which are achieved by selective gene delivery, specific gene expression or secretion of target proteins via genetic modification of autologous cells, dictate the outcome of apoptosis-based cancer gene therapy. Despite so far limited clinical success, gene therapy targeting the apoptotic machinery has great potential to benefit patients with threatening malignancies provided the availability of efficient and specific gene delivery and administration systems.
Collapse
Affiliation(s)
- Lin-Tao Jia
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China.
| | | | | |
Collapse
|