1
|
Chung C, Kudchodkar SB, Chung CN, Park YK, Xu Z, Pardi N, Abdel-Mohsen M, Muthumani K. Expanding the Reach of Monoclonal Antibodies: A Review of Synthetic Nucleic Acid Delivery in Immunotherapy. Antibodies (Basel) 2023; 12:46. [PMID: 37489368 PMCID: PMC10366852 DOI: 10.3390/antib12030046] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/26/2023] Open
Abstract
Harnessing the immune system to combat disease has revolutionized medical treatment. Monoclonal antibodies (mAbs), in particular, have emerged as important immunotherapeutic agents with clinical relevance in treating a wide range of diseases, including allergies, autoimmune diseases, neurodegenerative disorders, cancer, and infectious diseases. These mAbs are developed from naturally occurring antibodies and target specific epitopes of single molecules, minimizing off-target effects. Antibodies can also be designed to target particular pathogens or modulate immune function by activating or suppressing certain pathways. Despite their benefit for patients, the production and administration of monoclonal antibody therapeutics are laborious, costly, and time-consuming. Administration often requires inpatient stays and repeated dosing to maintain therapeutic levels, limiting their use in underserved populations and developing countries. Researchers are developing alternate methods to deliver monoclonal antibodies, including synthetic nucleic acid-based delivery, to overcome these limitations. These methods allow for in vivo production of monoclonal antibodies, which would significantly reduce costs and simplify administration logistics. This review explores new methods for monoclonal antibody delivery, including synthetic nucleic acids, and their potential to increase the accessibility and utility of life-saving treatments for several diseases.
Collapse
Affiliation(s)
| | | | - Curtis N Chung
- GeneOne Life Science, Inc., Seoul 04500, Republic of Korea
| | - Young K Park
- GeneOne Life Science, Inc., Seoul 04500, Republic of Korea
| | - Ziyang Xu
- Massachusetts General Hospital, Harvard University, Boston, MA 02114, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Kar Muthumani
- GeneOne Life Science, Inc., Seoul 04500, Republic of Korea
| |
Collapse
|
2
|
Joshi LR, Gálvez NM, Ghosh S, Weiner DB, Balazs AB. Delivery platforms for broadly neutralizing antibodies. Curr Opin HIV AIDS 2023; 18:191-208. [PMID: 37265268 PMCID: PMC10247185 DOI: 10.1097/coh.0000000000000803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
PURPOSE OF REVIEW Passive administration of broadly neutralizing antibodies (bNAbs) is being evaluated as a therapeutic approach to prevent or treat HIV infections. However, a number of challenges face the widespread implementation of passive transfer for HIV. To reduce the need of recurrent administrations of bNAbs, gene-based delivery approaches have been developed which overcome the limitations of passive transfer. RECENT FINDINGS The use of DNA and mRNA for the delivery of bNAbs has made significant progress. DNA-encoded monoclonal antibodies (DMAbs) have shown great promise in animal models of disease and the underlying DNA-based technology is now being tested in vaccine trials for a variety of indications. The COVID-19 pandemic greatly accelerated the development of mRNA-based technology to induce protective immunity. These advances are now being successfully applied to the delivery of monoclonal antibodies using mRNA in animal models. Delivery of bNAbs using viral vectors, primarily adeno-associated virus (AAV), has shown great promise in preclinical animal models and more recently in human studies. Most recently, advances in genome editing techniques have led to engineering of monoclonal antibody expression from B cells. These efforts aim to turn B cells into a source of evolving antibodies that can improve through repeated exposure to the respective antigen. SUMMARY The use of these different platforms for antibody delivery has been demonstrated across a wide range of animal models and disease indications, including HIV. Although each approach has unique strengths and weaknesses, additional advances in efficiency of gene delivery and reduced immunogenicity will be necessary to drive widespread implementation of these technologies. Considering the mounting clinical evidence of the potential of bNAbs for HIV treatment and prevention, overcoming the remaining technical challenges for gene-based bNAb delivery represents a relatively straightforward path towards practical interventions against HIV infection.
Collapse
Affiliation(s)
- Lok R. Joshi
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Nicolás M.S. Gálvez
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Sukanya Ghosh
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, PA 19104, USA
| | - David B. Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, PA 19104, USA
| | - Alejandro B. Balazs
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| |
Collapse
|
3
|
Andrews CD, Huang Y, Ho DD, Liberatore RA. In vivo expressed biologics for infectious disease prophylaxis: rapid delivery of DNA-based antiviral antibodies. Emerg Microbes Infect 2020; 9:1523-1533. [PMID: 32579067 PMCID: PMC7473320 DOI: 10.1080/22221751.2020.1787108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
With increasing frequency, humans are facing outbreaks of emerging infectious diseases (EIDs) with the potential to cause significant morbidity and mortality. In the most extreme instances, such outbreaks can become pandemics, as we are now witnessing with COVID-19. According to the World Health Organization, this new disease, caused by the novel coronavirus SARS-CoV-2, has already infected more than 10 million people worldwide and led to 499,913 deaths as of 29 June, 2020. How high these numbers will eventually go depends on many factors, including policies on travel and movement, availability of medical support, and, because there is no vaccine or highly effective treatment, the pace of biomedical research. Other than an approved antiviral drug that can be repurposed, monoclonal antibodies (mAbs) hold the most promise for providing a stopgap measure to lessen the impact of an outbreak while vaccines are in development. Technical advances in mAb identification, combined with the flexibility and clinical experience of mAbs in general, make them ideal candidates for rapid deployment. Furthermore, the development of mAb cocktails can provide a faster route to developing a robust medical intervention than searching for a single, outstanding mAb. In addition, mAbs are well-suited for integration into platform technologies for delivery, in which minimal components need to be changed in order to be redirected against a novel pathogen. In particular, utilizing the manufacturing and logistical benefits of DNA-based platform technologies in order to deliver one or more antiviral mAbs has the potential to revolutionize EID responses.
Collapse
Affiliation(s)
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, New York, NY, USA.,Columbia University Vagelos College of Physicans and Surgeons, New York, NY, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, New York, NY, USA.,Columbia University Vagelos College of Physicans and Surgeons, New York, NY, USA
| | | |
Collapse
|
4
|
Abstract
Antibody immunotherapy is revolutionizing modern medicine. The field has advanced dramatically over the past 40 years, driven in part by major advances in isolation and manufacturing technologies that have brought these important biologics to the forefront of modern medicine. However, the global uptake of monoclonal antibody (mAb) biologics is impeded by biophysical and biochemical liabilities, production limitations, the need for cold-chain storage and transport, as well as high costs of manufacturing and distribution. Some of these hurdles may be overcome through transient in vivo gene delivery platforms, such as non-viral synthetic plasmid DNA and messenger RNA vectors that are engineered to encode optimized mAb genes. These approaches turn the body into a biological factory for antibody production, eliminating many of the steps involved in bioprocesses and providing several other significant advantages, and differ from traditional gene therapy (permanent delivery) approaches. In this review, we focus on nucleic acid delivery of antibody employing synthetic plasmid DNA vector platforms, and RNA delivery, these being important approaches that are advancing simple, rapid, in vivo expression and having an impact in animal models of infectious diseases and cancer, among others.
Collapse
Affiliation(s)
- Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Mamadou A Bah
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
An engineered bispecific DNA-encoded IgG antibody protects against Pseudomonas aeruginosa in a pneumonia challenge model. Nat Commun 2017; 8:637. [PMID: 28935938 PMCID: PMC5608701 DOI: 10.1038/s41467-017-00576-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 07/10/2017] [Indexed: 01/19/2023] Open
Abstract
The impact of broad-spectrum antibiotics on antimicrobial resistance and disruption of the beneficial microbiome compels the urgent investigation of bacteria-specific approaches such as antibody-based strategies. Among these, DNA-delivered monoclonal antibodies (DMAbs), produced by muscle cells in vivo, potentially allow the prevention or treatment of bacterial infections circumventing some of the hurdles of protein IgG delivery. Here, we optimize DNA-delivered monoclonal antibodies consisting of two potent human IgG clones, including a non-natural bispecific IgG1 candidate, targeting Pseudomonas aeruginosa. The DNA-delivered monoclonal antibodies exhibit indistinguishable potency compared to bioprocessed IgG and protect against lethal pneumonia in mice. The DNA-delivered monoclonal antibodies decrease bacterial colonization of organs and exhibit enhanced adjunctive activity in combination with antibiotics. These studies support DNA-delivered monoclonal antibodies delivery as a potential strategy to augment the host immune response to prevent serious bacterial infections, and represent a significant advancement toward broader practical delivery of monoclonal antibody immunotherapeutics for additional infectious pathogens. DNA-delivered monoclonal antibodies (DMAbs) can be produced by muscle cells in vivo, potentially allowing prevention or treatment of infectious diseases. Here, the authors show that two DMAbs targeting Pseudomonas aeruginosa proteins confer protection against lethal pneumonia in mice.
Collapse
|
6
|
Andrews CD, Luo Y, Sun M, Yu J, Goff AJ, Glass PJ, Padte NN, Huang Y, Ho DD. In Vivo Production of Monoclonal Antibodies by Gene Transfer via Electroporation Protects against Lethal Influenza and Ebola Infections. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:74-82. [PMID: 29034261 PMCID: PMC5633264 DOI: 10.1016/j.omtm.2017.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/13/2017] [Indexed: 11/19/2022]
Abstract
Monoclonal antibodies (mAbs) have wide clinical utility, but global access is limited by high costs and impracticalities associated with repeated passive administration. Here, we describe an optimized electroporation-based DNA gene transfer platform technology that can be utilized for production of functional mAbs in vivo, with the potential to reduce costs and administration burdens. We demonstrate that multiple mAbs can be simultaneously expressed at protective concentrations for a protracted period of time using DNA doses and electroporation conditions that are feasible clinically. The expressed mAbs could also protect mice against lethal influenza or Ebola virus challenges. Our findings suggest that this DNA gene transfer platform technology could be a game-changing advance that expands access to effective mAb therapeutics globally.
Collapse
Affiliation(s)
- Chasity D. Andrews
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Yang Luo
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Ming Sun
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Jian Yu
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Arthur J. Goff
- US Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Pamela J. Glass
- US Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Neal N. Padte
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - Yaoxing Huang
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
| | - David D. Ho
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA
- Corresponding author: David D. Ho, Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA.
| |
Collapse
|
7
|
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: 47] [Impact Index Per Article: 5.9] [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
|
8
|
Rosazza C, Meglic SH, Zumbusch A, Rols MP, Miklavcic D. Gene Electrotransfer: A Mechanistic Perspective. Curr Gene Ther 2016; 16:98-129. [PMID: 27029943 PMCID: PMC5412002 DOI: 10.2174/1566523216666160331130040] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 11/22/2022]
Abstract
Gene electrotransfer is a powerful method of DNA delivery offering several medical applications, among the most promising of which are DNA vaccination and gene therapy for cancer treatment. Electroporation entails the application of electric fields to cells which then experience a local and transient change of membrane permeability. Although gene electrotransfer has been extensively studied in in vitro and in vivo environments, the mechanisms by which DNA enters and navigates through cells are not fully understood. Here we present a comprehensive review of the body of knowledge concerning gene electrotransfer that has been accumulated over the last three decades. For that purpose, after briefly reviewing the medical applications that gene electrotransfer can provide, we outline membrane electropermeabilization, a key process for the delivery of DNA and smaller molecules. Since gene electrotransfer is a multipart process, we proceed our review in describing step by step our current understanding, with particular emphasis on DNA internalization and intracellular trafficking. Finally, we turn our attention to in vivo testing and methodology for gene electrotransfer.
Collapse
Affiliation(s)
| | | | | | - Marie-Pierre Rols
- Institute of Pharmacology and Structural Biology (IPBS), CNRS UMR5089, 205 route de Narbonne, 31077 Toulouse, France.
| | | |
Collapse
|
9
|
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
|
10
|
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: 1.1] [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
|
11
|
Rochard A, Scherman D, Bigey P. Genetic immunization with plasmid DNA mediated by electrotransfer. Hum Gene Ther 2011; 22:789-98. [PMID: 21631165 DOI: 10.1089/hum.2011.092] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The concept of DNA immunization was first advanced in the early 1990s, but was not developed because of an initial lack of efficiency. Recent technical advances in plasmid design and gene delivery techniques have allowed renewed interest in the idea. Particularly, a better understanding of genetic immunization has led to construction of optimized plasmids and the use of efficient molecular adjuvants. The field also took great advantage of new delivery techniques such as electrotransfer. This is a simple physical technique consisting of injecting plasmid DNA into a target tissue and applying an electric field, allowing up to a thousandfold more expression of the transgene than naked DNA. DNA immunization mediated by electrotransfer is now effective in a variety of preclinical models against infectious or acquired diseases such as cancer or autoimmune diseases, and is making its way through the clinics in several ongoing phase I human clinical trials. This review will briefly describe genetic immunization mediated by electrotransfer and the main fields of application.
Collapse
Affiliation(s)
- Alice Rochard
- Unité de Pharmacologie Chimique et Génétique et d'Imagerie, CNRS, UMR8151, Paris, F-75006 France
| | | | | |
Collapse
|
12
|
The influence of skeletal muscle anisotropy on electroporation: in vivo study and numerical modeling. Med Biol Eng Comput 2010; 48:637-48. [PMID: 20424926 PMCID: PMC2886894 DOI: 10.1007/s11517-010-0614-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 04/11/2010] [Indexed: 11/30/2022]
Abstract
The aim of this study was to theoretically and experimentally investigate electroporation of mouse tibialis cranialis and to determine the reversible electroporation threshold values needed for parallel and perpendicular orientation of the applied electric field with respect to the muscle fibers. Our study was based on local electric field calculated with three-dimensional realistic numerical models, that we built, and in vivo visualization of electroporated muscle tissue. We established that electroporation of muscle cells in tissue depends on the orientation of the applied electric field; the local electric field threshold values were determined (pulse parameters: 8 × 100 μs, 1 Hz) to be 80 V/cm and 200 V/cm for parallel and perpendicular orientation, respectively. Our results could be useful electric field parameters in the control of skeletal muscle electroporation, which can be used in treatment planning of electroporation based therapies such as gene therapy, genetic vaccination, and electrochemotherapy.
Collapse
|
13
|
Alexandrenne C, Wijkhuisen A, Dkhissi F, Hanoux V, Priam F, Allard B, Boquet D, Couraud JY. Electrotransfer of cDNA Coding for a Heterologous Prion Protein Generates Autoantibodies Against Native Murine Prion Protein in Wild-Type Mice. DNA Cell Biol 2010; 29:121-31. [DOI: 10.1089/dna.2009.0940] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Coralie Alexandrenne
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
| | - Anne Wijkhuisen
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| | - Fatima Dkhissi
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| | - Vincent Hanoux
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| | - Fabienne Priam
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| | - Bertrand Allard
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
| | - Didier Boquet
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
| | - Jean-Yves Couraud
- CEA, iBiTecS, SPI, Laboratory of Antibody Engineering for Health, Gif sur Yvette, France
- UFR SdV, Paris Diderot University, Paris, France
| |
Collapse
|
14
|
Alexandrenne C, Wijkhuisen A, Dkhissi F, Hanoux V, Créminon C, Boquet D, Couraud JY. Generating antibodies against the native form of the human prion protein (hPrP) in wild-type animals: A comparison between DNA and protein immunizations. J Immunol Methods 2009; 341:41-9. [DOI: 10.1016/j.jim.2008.10.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 10/01/2008] [Accepted: 10/23/2008] [Indexed: 11/30/2022]
|
15
|
Samaranayake H, Wirth T, Schenkwein D, Räty JK, Ylä-Herttuala S. Challenges in monoclonal antibody-based therapies. Ann Med 2009; 41:322-31. [PMID: 19234897 DOI: 10.1080/07853890802698842] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are the fastest growing class of new therapeutic molecules. They hold great promises for the treatment of a variety of diseases, including chronic inflammatory diseases and cancer. However, the current manufacturing and purification processes cause limitations in the production capacity of therapeutic antibodies, leading to an increase in cost. Genetic delivery of therapeutic monoclonal antibodies by in vivo production offers a new potential solution to these problems. Firstly, therapeutic efficacy can be improved by maintaining stable therapeutic, non-toxic levels within the blood circulation over a long period of time. Repeated high-dose bolus injections could be avoided, thereby reducing the possibility of side-effects. Secondly, the high cost of manufacturing and purification of the therapeutic antibodies could be reduced, making an in vivo/ex vivo mAb gene transfer an economically viable and attractive option. In general, three approaches can be used for the stable long-term expression and secretion of therapeutic antibodies in vivo: 1) direct in vivo administration of integrating vectors carrying a mAb gene, 2) grafting of ex vivo genetically modified autologous cells, and 3) implantation of an encapsulated antibody producing heterologous or autologous cells. This paper describes the key factors and problems associated with the current antibody-based immunotherapies and reviews prospects for genetic in vivo delivery of therapeutic antibodies.
Collapse
|
16
|
Vigna E, Pacchiana G, Mazzone M, Chiriaco C, Fontani L, Basilico C, Pennacchietti S, Comoglio PM. "Active" cancer immunotherapy by anti-Met antibody gene transfer. Cancer Res 2008; 68:9176-83. [PMID: 19010889 DOI: 10.1158/0008-5472.can-08-1688] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gene therapy provides a still poorly explored opportunity to treat cancer by "active" immunotherapy as it enables the transfer of genes encoding antibodies directed against specific oncogenic proteins. By a bidirectional lentiviral vector, we transferred the cDNA encoding the heavy and light chains of a monoclonal anti-Met antibody (DN-30) to epithelial cancer cells. In vitro, the transduced cells synthesized and secreted correctly assembled antibodies with the expected high affinity, inducing down-regulation of the Met receptor and strong inhibition of the invasive growth response. The inhibitory activity resulted (a) from the interference of the antibody with the Met receptor intracellular processing ("cell autonomous activity," in cis) and (b) from the antibody-induced cleavage of Met expressed at the cell surface ("bystander effect," in trans). The monoclonal antibody gene transferred into live animals by systemic administration or by local intratumor delivery resulted in substantial inhibition of tumor growth. These data provide proof of concept both for targeting the Met receptor and for a gene transfer-based immunotherapy strategy.
Collapse
Affiliation(s)
- Elisa Vigna
- Laboratory for Gene Transfer and Therapy,Institute for Cancer Research and Treatment (IRCC), University of Turin Medical School, Turin, Italy
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Trollet C, Scherman D, Bigey P. Delivery of DNA into muscle for treating systemic diseases: advantages and challenges. Methods Mol Biol 2008; 423:199-214. [PMID: 18370200 DOI: 10.1007/978-1-59745-194-9_14] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
An efficient and safe method to deliver DNA in vivo is a requirement for several purposes, such as the study of gene function and gene therapy applications. Among the different nonviral delivery methods currently under investigation, in vivo DNA electrotransfer has proven to be one of the most efficient and simple methods. This technique is a physical method of gene delivery consisting of a local application of electric pulses after injection of DNA. This technique can be applied to almost any tissue of a living animal, including tumors, skin, liver, kidney, artery, retina, cornea, or even brain, but the focus of this review will be on electrotransfer of plasmid DNA into skeletal muscle and its possible therapeutic uses for systemic diseases. Skeletal muscle is a good target for electrotransfer of DNA because of the following features: a large volume of easily accessible tissue, an endocrine organ capable of expressing several local and systemic factors, and muscle fibers as postmitotic cells have a long lifespan, which allows long-term gene expression. In this review, we will describe the main characteristics of DNA electrotransfer, including toxicity and safety issues related to this technique. We will focus on the important possible therapeutic applications of electrotransfer for systemic diseases demonstrated in animal models in the recent years, in the fields of monogenic diseases, tissue-specific diseases, metabolic disorders, immune-system-related diseases, and cancer. Finally, we will discuss the advantages and challenges of this technique.
Collapse
Affiliation(s)
- Capucine Trollet
- Unité de Pharmacologie Chimique et Génétique, Faculté de Pharmacie, René Descartes Paris 5 University, Paris, France
| | | | | |
Collapse
|
18
|
Chen J, Su C, Lu Q, Shi W, Zhang Q, Wang X, Long J, Yang Q, Li L, Jia X, Wang J, Da W, Liu X, Wu M, Qian Q. Generation of adenovirus-mediated anti-CD20 antibody and its effect on B-cell deletion in mice and nonhuman primate cynomolgus monkey. Mol Cancer Ther 2008; 7:1562-8. [PMID: 18524844 DOI: 10.1158/1535-7163.mct-08-0297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jie Chen
- Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Avignolo C, Bagnasco L, Biasotti B, Melchiori A, Tomati V, Bauer I, Salis A, Chiossone L, Mingari MC, Orecchia P, Carnemolla B, Neri D, Zardi L, Parodi S. Internalization via Antennapedia protein transduction domain of an scFv antibody toward c-Myc protein. FASEB J 2007; 22:1237-45. [PMID: 18048579 DOI: 10.1096/fj.07-8865com] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We constructed a single-chain variable fragment miniantibody (G11-scFv) directed toward the transactivation domain of c-Myc, which is fused with the internalization domain Int of Antennapedia at its carboxyl terminus (a cargo-carrier construct). In ELISA experiments, an EC(50) for binding saturation was achieved at concentrations of G11-scFv-Int(-) of approximately 10(-8) M. Internalization of a fluoresceinated Fl-G11-scFv-Int(+) construct was observed in intact human cultured cells with confocal microscopy. After 5 h of incubation in medium containing 1 microM Fl-G11-scFv-Int(+) or Fl-G11-scFv-Int(-), fluorescence intensity was determined in individual cells, both for cytoplasmic and nuclear compartments: concentration levels of Fl-G11-scFv-Int(+), relative to the extracellular culture medium concentration, were 4-5 times higher in the cytoplasm, 7-8 times higher in the nucleus, and 10 times higher in the nucleoli. In the same experimental conditions, the Fl-G11-scFv-Int(-) construct was 3-4 times more concentrated outside of the cells than inside. Cell membranes kept their integrity after 5 h of incubation. The antiproliferative activity of our miniantibody was studied on HCT116 cells. Incubation with 4 microM G11-scFv-Int(+) for 4 days induced very significant statistical and biological growth inhibition, whereas Int alone was completely inactive. Miniantibodies capable of penetrating cell membranes dramatically broaden the potential for innovative therapeutic agents and attack of new targets.
Collapse
Affiliation(s)
- C Avignolo
- Department of Oncology, Biology and Genetics, University of Genoa, L. go R. Benzi 10, Genoa 16132, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hojman P, Gissel H, Gehl J. Sensitive and precise regulation of haemoglobin after gene transfer of erythropoietin to muscle tissue using electroporation. Gene Ther 2007; 14:950-9. [PMID: 17410179 DOI: 10.1038/sj.gt.3302951] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Electroporation-based gene transfer (electro gene transfer (EGT)) is gaining increasing momentum, in particular for muscle tissue, where long-term high-level expression is obtainable. Induction of expression using the Tet-On system was previously established; however, attempts to reach a predefined target dose - a prescription, have not been reported. We set three target haemoglobin levels (10, 12 and 14 mmol/l, base level was 8.2 mmol/l) and aimed at them by transferring the erythropoietin (EPO) gene to mouse tibialis cranialis (TC) muscle, and varying (1) DNA amount, (2) muscle mass transfected and (3) induction with the Tet-On system. Results showed that (a) using GFP, luciferase and EPO low DNA amounts were needed. In fact, 0.5 microg of DNA to one TC muscle led to significant Hgb elevation - this amount extrapolates to 1.4 mg of DNA in humans, (b) three prescribers hit the targets with average Hgb of 10.5, 12.0 and 13.7 mmol/l, (c) different approaches could be used, (d) undershooting could be corrected by retransferring, and (e) overshooting could be alleviated by reducing dose of inducer (doxycycline (dox)). In conclusion, this study shows that using EGT to muscle, a preset level of protein expression can be reached. This is of great interest for future clinical use.
Collapse
Affiliation(s)
- P Hojman
- Laboratory of the Department of Oncology, University of Copenhagen at Herlev Hospital, Herlev, Denmark
| | | | | |
Collapse
|
21
|
Affiliation(s)
- Loree C Heller
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | | |
Collapse
|
22
|
Abstract
The utilisation of nonviral gene delivery methods has been increasing steadily, however, a drawback has been the relative low efficiency of gene transfer with naked DNA compared with viral delivery methods. In vivo electroporation, which has previously been used clinically to deliver chemotherapeutic agents, also enhances the delivery of plasmid DNA and has been used to deliver plasmids to several tissue types, particularly muscle and tumour. Recently, a large number of preclinical studies for a variety of therapeutic modalities have demonstrated the potential of electrically mediated gene transfer. Although clinical trials using gene transfer with in vivo electroporation have not as yet been realised, the tremendous growth of this technology suggests that the first trials will soon be initiated.
Collapse
MESH Headings
- Animals
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/therapy
- Electroporation
- Erythropoietin/administration & dosage
- Erythropoietin/genetics
- Erythropoietin/metabolism
- Forecasting
- Gene Expression Regulation
- Gene Transfer Techniques
- Genetic Therapy/methods
- Genetic Therapy/trends
- Growth Substances/administration & dosage
- Growth Substances/genetics
- Growth Substances/metabolism
- Hematologic Diseases/genetics
- Hematologic Diseases/metabolism
- Hematologic Diseases/therapy
- Humans
- Injections, Intramuscular
- Interleukin-12/administration & dosage
- Interleukin-12/genetics
- Interleukin-12/metabolism
- Melanoma/genetics
- Melanoma/metabolism
- Melanoma/therapy
- Plasmids/administration & dosage
- Plasmids/genetics
- Plasmids/metabolism
- Protein Deficiency/genetics
- Protein Deficiency/metabolism
- Protein Deficiency/therapy
- Toxins, Biological/administration & dosage
- Toxins, Biological/genetics
- Toxins, Biological/metabolism
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/metabolism
Collapse
Affiliation(s)
- Loree C Heller
- University of South Florida, Center of Molecular Delivery, Department of Medical Microbiology and Immunology, Center for Molecular Delivery, College of Medicine, Tampa, 33612-4799, USA.
| | | | | |
Collapse
|
23
|
|
24
|
Bakker JM, Bleeker WK, Parren PWHI. Therapeutic antibody gene transfer: an active approach to passive immunity. Mol Ther 2005; 10:411-6. [PMID: 15336642 DOI: 10.1016/j.ymthe.2004.06.865] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2004] [Revised: 06/18/2004] [Accepted: 06/18/2004] [Indexed: 01/08/2023] Open
Abstract
Advances in gene transfer approaches are enabling the possibility of applying therapeutic antibodies using DNA. In particular gene transfer in combination with electroporation is promising and can result in generating in vivo antibody concentrations in the low therapeutic range. However, several important problems need to be dealt with before antibody gene transfer can become a valuable supplement to the current therapies. As antibody production following gene transfer is difficult to control, the danger of inducing autoimmune conditions or uncontrollable side effects occurs in cases in which autologous antigens are targeted. It is suggested that the most promising area of application therefore appears to be infectious disease in which heterologous antigens are targeted and concerns for long-term antibody exposure are minimal. Finally, genes encoding fully human antibodies will enhance long-term expression and decrease problems linked to immunogenicity.
Collapse
Affiliation(s)
- Joost M Bakker
- Genmab B.V., Yalelaan 60, P.O. Box 85199, 3508 AD Utrecht, The Netherlands
| | | | | |
Collapse
|
25
|
Fang J, Qian JJ, Yi S, Harding TC, Tu GH, VanRoey M, Jooss K. Stable antibody expression at therapeutic levels using the 2A peptide. Nat Biotechnol 2005; 23:584-90. [PMID: 15834403 DOI: 10.1038/nbt1087] [Citation(s) in RCA: 306] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2005] [Accepted: 03/10/2005] [Indexed: 11/09/2022]
Abstract
Therapeutic monoclonal antibodies (mAbs) are currently being developed for the treatment of cancer and other diseases. Despite clinical success, widespread application of mAb therapies may be limited by manufacturing capabilities. In this paper, we describe a mAb delivery system that allows continuous production of a full-length antibody at high-concentrations in vivo after gene transfer. The mAb is expressed from a single open reading frame by linking the heavy and light chains with a 2A self-processing peptide derived from the foot-and-mouth disease virus. Using this expression system, we generated a recombinant adeno-associated virus vector encoding the VEGFR2-neutralizing mAb DC101 (rAAV8-DC101). A single dose of rAAV8-DC101 resulted in long-term expression of >1,000 microg/ml of DC101 in mice, demonstrating significant anti-tumor efficacy. This report describes the first feasible gene therapy approach for stable delivery of mAbs at therapeutic levels, which may serve as an attractive alternative to direct injection of mAbs.
Collapse
Affiliation(s)
- Jianmin Fang
- Department of Preclinical Oncology and Immunology, Cell Genesys, Inc., 500 Forbes Blvd., S. San Francisco, California 94080, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
Mir LM, Moller PH, André F, Gehl J. Electric pulse-mediated gene delivery to various animal tissues. ADVANCES IN GENETICS 2005; 54:83-114. [PMID: 16096009 DOI: 10.1016/s0065-2660(05)54005-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Electroporation designates the use of electric pulses to transiently permeabilize the cell membrane. It has been shown that DNA can be transferred to cells through a combined effect of electric pulses causing (1) permeabilization of the cell membrane and (2) an electrophoretic effect on DNA, leading the polyanionic molecule to move toward or across the destabilized membrane. This process is now referred to as DNA electrotransfer or electro gene transfer (EGT). Several studies have shown that EGT can be highly efficient, with low variability both in vitro and in vivo. Furthermore, the area transfected is restricted by the placement of the electrodes, and is thus highly controllable. This has led to an increasing use of the technology to transfer reporter or therapeutic genes to various tissues, as evidenced from the large amount of data accumulated on this new approach for non-viral gene therapy, termed electrogenetherapy (EGT as well). By transfecting cells with a long lifetime, such as muscle fibers, a very long-term expression of genes can be obtained. A great variety of tissues have been transfected successfully, from muscle as the most extensively used, to both soft (e.g., spleen) and hard tissue (e.g., cartilage). It has been shown that therapeutic levels of systemically circulating proteins can be obtained, opening possibilities for using EGT therapeutically. This chapter describes the various aspects of in vivo gene delivery by means of electric pulses, from important issues in methodology to updated results concerning the electrotransfer of reporter and therapeutic genes to different tissues.
Collapse
Affiliation(s)
- Lluis M Mir
- Laboratory of Vectorology and Gene Transfer, UMR 8121 CNRS Institut Gustave-Roussy, F-94805 Villejuif Cédex, France
| | | | | | | |
Collapse
|
27
|
André F, Mir LM. DNA electrotransfer: its principles and an updated review of its therapeutic applications. Gene Ther 2004; 11 Suppl 1:S33-42. [PMID: 15454955 DOI: 10.1038/sj.gt.3302367] [Citation(s) in RCA: 181] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The use of electric pulses to transfect all types of cells is well known and regularly used in vitro for bacteria and eukaryotic cells transformation. Electric pulses can also be delivered in vivo either transcutaneously or with electrodes in direct contact with the tissues. After injection of naked DNA in a tissue, appropriate local electric pulses can result in a very high expression of the transferred genes. This manuscript describes the evolution in the concepts and the various optimization steps that have led to the use of combinations of pulses that fit with the known roles of the electric pulses in DNA electrotransfer, namely cell electropermeabilization and DNA electrophoresis. A summary of the main applications published until now is also reported, restricted to the in vivo preclinical trials using therapeutic genes.
Collapse
Affiliation(s)
- F André
- Laboratory of Vectorology and Gene Transfer, UMR 8121 CNRS - Institut Gustave-Roussy, Villejuif Cedex, France
| | | |
Collapse
|
28
|
Abstract
The idea of enhancing or establishing effective immune response against endogenously developed tumor cells is not novel. More than a hundred years ago, bacterial components were used to develop antitumor immune response. Later, when a number of immune system-effecting cytokines had been discovered, they were used for systemic treatment of cancer patients. However, systemic treatment often resulted in even negative outcome. Recent developments of genetic approaches of cell modifications allowed developing of modern techniques of targeted tumor cell elimination. In the present paper, we review modern trends of the antitumor response enhancement based on immunoregulatory gene transfer into different cell types both in vivo and in vitro. Almost all these approaches are based on the activation of the adaptive arm of the immune system in response to tumor cells. However, recent studies indicate that the innate arm of the immune system, as well as adaptive arm, is involved in tumor suppression. The innate immune system uses nonrearranging germline receptors, which could trigger cellular effector responses that are conditional (or instructive) to the subsequent adaptive immune response. Last years' viewpoints on 'self' and 'non-self' recognition and primary induction of the immune response have changed. The key role of lymphocytes is pathogen recognition and, following immune response induction, switched on the central role of dendritic cells in 'non-self' recognition and induction of both innate and adaptive responses. Moreover, innate response is supposed to be an essential starting point in induction of successful and effective acquired response. Most cancer vaccines do not have 'non-self' marks presentation due to their endogenous origin, thus lacking their effectiveness in the induction of the specific long-lasting immune response. Taking this point into consideration, we can conclude that to make cancer vaccine more effective we have to present tumor antigens, together with the molecules that can potentially activate downstream 'non-self' recognition events not in parallel, but as a consequence of tumor antigen processing and presentation.
Collapse
Affiliation(s)
- S S Larin
- Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
| | | | | |
Collapse
|