1
|
Muravyeva A, Smirnikhina S. Adenoviral Vectors for Gene Therapy of Hereditary Diseases. BIOLOGY 2024; 13:1052. [PMID: 39765719 PMCID: PMC11673936 DOI: 10.3390/biology13121052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
Adenoviral vectors (AdVs) are effective vectors for gene therapy due to their broad tropism, high capacity, and high transduction efficiency, which makes them actively used as oncolytic vectors and for creating vector vaccines. However, despite their numerous advantages, AdVs have not yet found their place in gene therapy for hereditary diseases. This review provides an overview of AdVs, their features, and clinical trials using them for gene replacement therapy in monogenic diseases and analyzes the reasons for the failures of these studies. Additionally, current research on the modification of AdVs to reduce immune responses and target delivery is discussed.
Collapse
Affiliation(s)
| | - Svetlana Smirnikhina
- Laboratory of Genome Editing, Research Centre for Medical Genetics, Moskvorechye, 1, 115522 Moscow, Russia
| |
Collapse
|
2
|
Ingusci S, Hall BL, Goins WF, Cohen JB, Glorioso JC. Viral vectors for gene delivery to the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:59-81. [PMID: 39341663 DOI: 10.1016/b978-0-323-90120-8.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Brain diseases with a known or suspected genetic basis represent an important frontier for advanced therapeutics. The central nervous system (CNS) is an intricate network in which diverse cell types with multiple functions communicate via complex signaling pathways, making therapeutic intervention in brain-related diseases challenging. Nevertheless, as more information on the molecular genetics of brain-related diseases becomes available, genetic intervention using gene therapeutic strategies should become more feasible. There remain, however, several significant hurdles to overcome that relate to (i) the development of appropriate gene vectors and (ii) methods to achieve local or broad vector delivery. Clearly, gene delivery tools must be engineered for distribution to the correct cell type in a specific brain region and to accomplish therapeutic transgene expression at an appropriate level and duration. They also must avoid all toxicity, including the induction of inflammatory responses. Over the last 40 years, various types of viral vectors have been developed as tools to introduce therapeutic genes into the brain, primarily targeting neurons. This review describes the most prominent vector systems currently approaching clinical application for CNS disorders and highlights both remaining challenges as well as improvements in vector designs that achieve greater safety, defined tropism, and therapeutic gene expression.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bonnie L Hall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
3
|
Minskaia E, Galieva A, Egorov AD, Ivanov R, Karabelsky A. Viral Vectors in Gene Replacement Therapy. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:2157-2178. [PMID: 38462459 DOI: 10.1134/s0006297923120179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/29/2023] [Accepted: 10/17/2023] [Indexed: 03/12/2024]
Abstract
Throughout the years, several hundred million people with rare genetic disorders have been receiving only symptom management therapy. However, research and development efforts worldwide have led to the development of long-lasting, highly efficient, and safe gene therapy for a wide range of hereditary diseases. Improved viral vectors are now able to evade the preexisting immunity and more efficiently target and transduce therapeutically relevant cells, ensuring genome maintenance and expression of transgenes at the relevant levels. Hematological, ophthalmological, neurodegenerative, and metabolic therapeutic areas have witnessed successful treatment of hemophilia and muscular dystrophy, restoration of immune system in children with immunodeficiencies, and restoration of vision. This review focuses on three leading vector platforms of the past two decades: adeno-associated viruses (AAVs), adenoviruses (AdVs), and lentiviruses (LVs). Special attention is given to successful preclinical and clinical studies that have led to the approval of gene therapies: six AAV-based (Glybera® for lipoprotein lipase deficiency, Luxturna® for retinal dystrophy, Zolgensma® for spinal muscular atrophy, Upstaza® for AADC, Roctavian® for hemophilia A, and Hemgenix® for hemophilia B) and three LV-based (Libmeldy® for infantile metachromatic leukodystrophy, Zynteglo® for β-thalassemia, and Skysona® for ALD). The review also discusses the problems that arise in the development of gene therapy treatments, which, nevertheless, do not overshadow the successes of already developed gene therapies and the hope these treatments give to long-suffering patients and their families.
Collapse
Affiliation(s)
- Ekaterina Minskaia
- Scientific Center of Translational Medicine, Department of Gene Therapy, Sirius University of Science and Technology, Sochi, 354530, Russia.
| | - Alima Galieva
- Scientific Center of Translational Medicine, Department of Gene Therapy, Sirius University of Science and Technology, Sochi, 354530, Russia
| | - Alexander D Egorov
- Scientific Center of Translational Medicine, Department of Gene Therapy, Sirius University of Science and Technology, Sochi, 354530, Russia
| | - Roman Ivanov
- Scientific Center of Translational Medicine, Department of Gene Therapy, Sirius University of Science and Technology, Sochi, 354530, Russia
| | - Alexander Karabelsky
- Scientific Center of Translational Medicine, Department of Gene Therapy, Sirius University of Science and Technology, Sochi, 354530, Russia
| |
Collapse
|
4
|
Thampi P, Samulski RJ, Grieger JC, Phillips JN, McIlwraith CW, Goodrich LR. Gene therapy approaches for equine osteoarthritis. Front Vet Sci 2022; 9:962898. [PMID: 36246316 PMCID: PMC9558289 DOI: 10.3389/fvets.2022.962898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/08/2022] [Indexed: 01/24/2023] Open
Abstract
With an intrinsically low ability for self-repair, articular cartilage injuries often progress to cartilage loss and joint degeneration resulting in osteoarthritis (OA). Osteoarthritis and the associated articular cartilage changes can be debilitating, resulting in lameness and functional disability both in human and equine patients. While articular cartilage damage plays a central role in the pathogenesis of OA, the contribution of other joint tissues to the pathogenesis of OA has increasingly been recognized thus prompting a whole organ approach for therapeutic strategies. Gene therapy methods have generated significant interest in OA therapy in recent years. These utilize viral or non-viral vectors to deliver therapeutic molecules directly into the joint space with the goal of reprogramming the cells' machinery to secrete high levels of the target protein at the site of injection. Several viral vector-based approaches have demonstrated successful gene transfer with persistent therapeutic levels of transgene expression in the equine joint. As an experimental model, horses represent the pathology of human OA more accurately compared to other animal models. The anatomical and biomechanical similarities between equine and human joints also allow for the use of similar imaging and diagnostic methods as used in humans. In addition, horses experience naturally occurring OA and undergo similar therapies as human patients and, therefore, are a clinically relevant patient population. Thus, further studies utilizing this equine model would not only help advance the field of human OA therapy but also benefit the clinical equine patients with naturally occurring joint disease. In this review, we discuss the advancements in gene therapeutic approaches for the treatment of OA with the horse as a relevant patient population as well as an effective and commonly utilized species as a translational model.
Collapse
Affiliation(s)
- Parvathy Thampi
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Joshua C. Grieger
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC, United States
| | - Jennifer N. Phillips
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - C. Wayne McIlwraith
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States
| | - Laurie R. Goodrich
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Research Institute, College of Veterinary Medicine, Colorado State University, Fort Collins, CO, United States,*Correspondence: Laurie R. Goodrich
| |
Collapse
|
5
|
Rice A, Verma M, Voigt E, Battisti P, Beaver S, Reed S, Dinkins K, Mody S, Zakin L, Tanaka S, Morimoto B, Olson CA, Gabitzsch E, Safrit JT, Spilman P, Casper C, Soon-Shiong P. Heterologous saRNA Prime, DNA Dual-Antigen Boost SARS-CoV-2 Vaccination Elicits Robust Cellular Immunogenicity and Cross-Variant Neutralizing Antibodies. Front Immunol 2022; 13:910136. [PMID: 35911728 PMCID: PMC9335885 DOI: 10.3389/fimmu.2022.910136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
Abstract
We assessed if immune responses are enhanced in CD-1 mice by heterologous vaccination with two different nucleic acid-based COVID-19 vaccines: a next-generation human adenovirus serotype 5 (hAd5)-vectored dual-antigen spike (S) and nucleocapsid (N) vaccine (AdS+N) and a self-amplifying and -adjuvanted S RNA vaccine (AAHI-SC2) delivered by a nanostructured lipid carrier. The AdS+N vaccine encodes S modified with a fusion motif to increase cell-surface expression and an N antigen modified with an Enhanced T-cell Stimulation Domain (N-ETSD) to direct N to the endosomal/lysosomal compartment and increase MHC class I and II stimulation potential. The S sequence in the AAHI-SC2 vaccine comprises the D614G mutation, two prolines to stabilize S in the prefusion conformation, and 3 glutamines in the furin cleavage region to confer protease resistance. CD-1 mice received vaccination by homologous and heterologous prime > boost combinations. Humoral responses to S were the highest with any regimen that included the AAHI-SC2 vaccine, and IgG bound to wild type and Delta (B.1.617.2) variant S1 at similar levels. An AAHI-SC2 prime followed by an AdS+N boost particularly enhanced CD4+ and CD8+ T-cell responses to both wild type and Delta S peptides relative to all other vaccine regimens. Sera from mice receiving AAHI-SC2 homologous or heterologous vaccination were found to be highly neutralizing for all pseudovirus strains tested: Wuhan, Beta, Delta, and Omicron strains. The findings here, taken in consideration with the availability of both vaccines in thermostable formulations, support the testing of heterologous vaccination by an AAHI-SC2 > AdS+N regimen in animal models of SARS-CoV-2 infection to assess its potential to provide increased protection against emerging SARS-CoV-2 variants particularly in regions of the world where the need for cold-chain storage has limited the distribution of other vaccines.
Collapse
Affiliation(s)
- Adrian Rice
- ImmunityBio, Inc., Culver City, CA, United States
| | - Mohit Verma
- ImmunityBio, Inc., Culver City, CA, United States
| | - Emily Voigt
- Access to Advanced Health Institute (AAHI), Seattle, WA, United States
| | - Peter Battisti
- Access to Advanced Health Institute (AAHI), Seattle, WA, United States
| | - Sam Beaver
- Access to Advanced Health Institute (AAHI), Seattle, WA, United States
| | - Sierra Reed
- Access to Advanced Health Institute (AAHI), Seattle, WA, United States
| | - Kyle Dinkins
- ImmunityBio, Inc., Culver City, CA, United States
| | - Shivani Mody
- ImmunityBio, Inc., Culver City, CA, United States
| | - Lise Zakin
- ImmunityBio, Inc., Culver City, CA, United States
| | - Shiho Tanaka
- ImmunityBio, Inc., Culver City, CA, United States
| | | | | | | | | | | | - Corey Casper
- Access to Advanced Health Institute (AAHI), Seattle, WA, United States
- Departments of Medicine and Global Health, University of Washington, Seattle, WA, United States
| | | |
Collapse
|
6
|
Zhao Z, Anselmo AC, Mitragotri S. Viral vector-based gene therapies in the clinic. Bioeng Transl Med 2022; 7:e10258. [PMID: 35079633 PMCID: PMC8780015 DOI: 10.1002/btm2.10258] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
Gene therapies are currently one of the most investigated therapeutic modalities in both the preclinical and clinical settings and have shown promise in treating a diverse spectrum of diseases. Gene therapies aim at introducing a gene material in target cells and represent a promising approach to cure diseases that were thought to be incurable by conventional modalities. In many cases, a gene therapy requires a vector to deliver gene therapeutics into target cells; viral vectors are among the most widely studied vectors owing to their distinguished advantages such as outstanding transduction efficiency. With decades of development, viral vector-based gene therapies have achieved promising clinical outcomes with many products approved for treating a range of diseases including cancer, infectious diseases and monogenic diseases. In addition, a number of active clinical trials are underway to further expand their therapeutic potential. In this review, we highlight the diversity of viral vectors, review approved products, and discuss the current clinical landscape of in vivo viral vector-based gene therapies. We have reviewed 13 approved products and their clinical applications. We have also analyzed more than 200 active trials based on various viral vectors and discussed their respective therapeutic applications. Moreover, we provide a critical analysis of the major translational challenges for in vivo viral vector-based gene therapies and discuss possible strategies to address the same.
Collapse
Affiliation(s)
- Zongmin Zhao
- Department of Pharmaceutical Sciences, College of PharmacyUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Aaron C. Anselmo
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| |
Collapse
|
7
|
Gabitzsch E, Safrit JT, Verma M, Rice A, Sieling P, Zakin L, Shin A, Morimoto B, Adisetiyo H, Wong R, Bezawada A, Dinkins K, Balint J, Peykov V, Garban H, Liu P, Bacon A, Bone P, Drew J, Sanford DC, Spilman P, Sender L, Rabizadeh S, Niazi K, Soon-Shiong P. Dual-Antigen COVID-19 Vaccine Subcutaneous Prime Delivery With Oral Boosts Protects NHP Against SARS-CoV-2 Challenge. Front Immunol 2021; 12:729837. [PMID: 34603305 PMCID: PMC8481919 DOI: 10.3389/fimmu.2021.729837] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/20/2021] [Indexed: 12/25/2022] Open
Abstract
We have developed a dual-antigen COVID-19 vaccine incorporating genes for a modified SARS-CoV-2 spike protein (S-Fusion) and the viral nucleocapsid (N) protein with an Enhanced T-cell Stimulation Domain (N-ETSD) to increase the potential for MHC class II responses. The vaccine antigens are delivered by a human adenovirus serotype 5 platform, hAd5 [E1-, E2b-, E3-], previously demonstrated to be effective in the presence of Ad immunity. Vaccination of rhesus macaques with the hAd5 S-Fusion + N-ETSD vaccine by subcutaneous prime injection followed by two oral boosts elicited neutralizing anti-S IgG and T helper cell 1-biased T-cell responses to both S and N that protected the upper and lower respiratory tracts from high titer (1 x 106 TCID50) SARS-CoV-2 challenge. Notably, viral replication was inhibited within 24 hours of challenge in both lung and nasal passages, becoming undetectable within 7 days post-challenge.
Collapse
Affiliation(s)
| | | | - Mohit Verma
- ImmunityBio, Inc., Culver City, CA, United States
| | - Adrian Rice
- ImmunityBio, Inc., Culver City, CA, United States
| | | | - Lise Zakin
- ImmunityBio, Inc., Culver City, CA, United States
| | - Annie Shin
- ImmunityBio, Inc., Culver City, CA, United States
| | | | | | - Raymond Wong
- ImmunityBio, Inc., Culver City, CA, United States
| | | | - Kyle Dinkins
- ImmunityBio, Inc., Culver City, CA, United States
| | | | | | | | - Philip Liu
- ImmunityBio, Inc., Culver City, CA, United States
| | | | - Pete Bone
- IosBio, Burgess Hill, United Kingdom
| | - Jeff Drew
- IosBio, Burgess Hill, United Kingdom
| | | | | | | | | | - Kayvan Niazi
- ImmunityBio, Inc., Culver City, CA, United States
| | | |
Collapse
|
8
|
Okamura S, Ebina H. Could live attenuated vaccines better control COVID-19? Vaccine 2021; 39:5719-5726. [PMID: 34426024 PMCID: PMC8354792 DOI: 10.1016/j.vaccine.2021.08.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/23/2022]
Abstract
In an effort to control the COVID-19 pandemic, large-scale vaccination is being implemented in various countries using anti-SARS-CoV-2 vaccines based on mRNAs, adenovirus vectors, and inactivated viruses. However, there are concerns regarding adverse effects, such as the induction of fever attributed to mRNA vaccines and pre-existing immunity against adenovirus vectored vaccines or their possible involvement in the development of thrombosis. The induction of antibodies against the adenovirus vector itself constitutes another hindrance, rendering boosting vaccinations ineffective. Additionally, it has been questioned whether inactivated vaccines that predominantly induce humoral immunity are effective against newly arising variants, as some isolated strains were found to be resistant to the serum from COVID-19-recovered patients. Although the number of vaccinated people is steadily increasing on a global scale, it is still necessary to develop vaccines to address the difficulties and concerns mentioned above. Among the various vaccine modalities, live attenuated vaccines have been considered the most effective, since they closely replicate a natural infection without the burden of the disease. In our attempt to provide an additional option to the repertoire of COVID-19 vaccines, we succeeded in isolating temperature-sensitive strains with unique phenotypes that could serve as seeds for a live attenuated vaccine. In this review article, we summarize the characteristics of the currently approved SARS-CoV-2 vaccines and discuss their advantages and disadvantages. In particular, we focus on the novel temperature-sensitive variants of SARS-CoV-2 that we have recently isolated, and their potential application as live-attenuated vaccines. Based on a thorough evaluation of the different vaccine modalities, we argue that it is important to optimize usage not only based on efficacy, but also on the phases of the pandemic. Our findings can be used to inform vaccination practices and improve global recovery from the COVID-19 pandemic.
Collapse
Affiliation(s)
- Shinya Okamura
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Hirotaka Ebina
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan; The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan; Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
9
|
Rice A, Verma M, Shin A, Zakin L, Sieling P, Tanaka S, Balint J, Dinkins K, Adisetiyo H, Morimoto B, Higashide W, Anders Olson C, Mody S, Spilman P, Gabitzsch E, Safrit JT, Rabizadeh S, Niazi K, Soon-Shiong P. Intranasal plus subcutaneous prime vaccination with a dual antigen COVID-19 vaccine elicits T-cell and antibody responses in mice. Sci Rep 2021; 11:14917. [PMID: 34290317 PMCID: PMC8295250 DOI: 10.1038/s41598-021-94364-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022] Open
Abstract
We have developed a COVID-19 vaccine, hAd5 S-Fusion + N-ETSD, that expresses SARS-CoV-2 spike (S) and nucleocapsid (N) proteins with modifications to increase immune responses delivered using a human adenovirus serotype 5 (hAd5) platform. Here, we demonstrate subcutaneous (SC) prime and SC boost vaccination of CD-1 mice with this dual-antigen vaccine elicits T-helper cell 1 (Th1) biased T-cell and humoral responses to both S and N that are greater than those seen with hAd5 S wild type delivering only unmodified S. We then compared SC to intranasal (IN) prime vaccination with SC or IN boosts and show that an IN prime with an IN boost is as effective at generating Th1 biased humoral responses as the other combinations tested, but an SC prime with an IN or SC boost elicits greater T cell responses. Finally, we used a combined SC plus IN (SC + IN) prime with or without a boost and found the SC + IN prime alone to be as effective in generating humoral and T-cell responses as the SC + IN prime with a boost. The finding that SC + IN prime-only delivery has the potential to provide broad immunity-including mucosal immunity-against SARS-CoV-2 supports further testing of this vaccine and delivery approach in animal models of viral challenge.
Collapse
Affiliation(s)
- Adrian Rice
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Mohit Verma
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Annie Shin
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Lise Zakin
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Peter Sieling
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Shiho Tanaka
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Joseph Balint
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Kyle Dinkins
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Helty Adisetiyo
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Brett Morimoto
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Wendy Higashide
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - C Anders Olson
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Shivani Mody
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | - Patricia Spilman
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | | | - Jeffrey T Safrit
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | | | - Kayvan Niazi
- ImmunityBio, Inc., 9920 Jefferson Blvd, Culver City, CA, 90232, USA
| | | |
Collapse
|
10
|
Capsid and Genome Modification Strategies to Reduce the Immunogenicity of Adenoviral Vectors. Int J Mol Sci 2021; 22:ijms22052417. [PMID: 33670859 PMCID: PMC7957472 DOI: 10.3390/ijms22052417] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 01/01/2023] Open
Abstract
Adenovirus-based gene transfer vectors are the most frequently used vector type in gene therapy clinical trials to date, and they play an important role as genetic vaccine candidates during the ongoing SARS-CoV-2 pandemic. Immediately upon delivery, adenovirus-based vectors exhibit multiple complex vector-host interactions and induce innate and adaptive immune responses. This can severely limit their safety and efficacy, particularly after delivery through the blood stream. In this review article we summarize two strategies to modulate Ad vector-induced immune responses: extensive genomic and chemical capsid modifications. Both strategies have shown beneficial effects in a number of preclinical studies while potential synergistic effects warrant further investigations.
Collapse
|
11
|
Bulcha JT, Wang Y, Ma H, Tai PWL, Gao G. Viral vector platforms within the gene therapy landscape. Signal Transduct Target Ther 2021; 6:53. [PMID: 33558455 PMCID: PMC7868676 DOI: 10.1038/s41392-021-00487-6] [Citation(s) in RCA: 680] [Impact Index Per Article: 170.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/05/2020] [Accepted: 10/23/2020] [Indexed: 01/30/2023] Open
Abstract
Throughout its 40-year history, the field of gene therapy has been marked by many transitions. It has seen great strides in combating human disease, has given hope to patients and families with limited treatment options, but has also been subject to many setbacks. Treatment of patients with this class of investigational drugs has resulted in severe adverse effects and, even in rare cases, death. At the heart of this dichotomous field are the viral-based vectors, the delivery vehicles that have allowed researchers and clinicians to develop powerful drug platforms, and have radically changed the face of medicine. Within the past 5 years, the gene therapy field has seen a wave of drugs based on viral vectors that have gained regulatory approval that come in a variety of designs and purposes. These modalities range from vector-based cancer therapies, to treating monogenic diseases with life-altering outcomes. At present, the three key vector strategies are based on adenoviruses, adeno-associated viruses, and lentiviruses. They have led the way in preclinical and clinical successes in the past two decades. However, despite these successes, many challenges still limit these approaches from attaining their full potential. To review the viral vector-based gene therapy landscape, we focus on these three highly regarded vector platforms and describe mechanisms of action and their roles in treating human disease.
Collapse
Affiliation(s)
- Jote T Bulcha
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic medical sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Hong Ma
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA.
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
12
|
Sena-Esteves M, Gao G. Introducing Genes into Mammalian Cells: Viral Vectors. Cold Spring Harb Protoc 2020; 2020:095513. [PMID: 32457039 DOI: 10.1101/pdb.top095513] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Over the years, many different viral vector systems have been developed to take advantage of the specific biological properties and tropisms of a large number of mammalian viruses. As a result, researchers wanting to introduce and/or express genes in mammalian cells have many options, as discussed here.
Collapse
|
13
|
Kleinlogel S, Vogl C, Jeschke M, Neef J, Moser T. Emerging approaches for restoration of hearing and vision. Physiol Rev 2020; 100:1467-1525. [DOI: 10.1152/physrev.00035.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Impairments of vision and hearing are highly prevalent conditions limiting the quality of life and presenting a major socioeconomic burden. For long, retinal and cochlear disorders have remained intractable for causal therapies, with sensory rehabilitation limited to glasses, hearing aids, and electrical cochlear or retinal implants. Recently, the application of gene therapy and optogenetics to eye and ear has generated hope for a fundamental improvement of vision and hearing restoration. To date, one gene therapy for the restoration of vision has been approved and undergoing clinical trials will broaden its application including gene replacement, genome editing, and regenerative approaches. Moreover, optogenetics, i.e. controlling the activity of cells by light, offers a more general alternative strategy. Over little more than a decade, optogenetic approaches have been developed and applied to better understand the function of biological systems, while protein engineers have identified and designed new opsin variants with desired physiological features. Considering potential clinical applications of optogenetics, the spotlight is on the sensory systems. Multiple efforts have been undertaken to restore lost or hampered function in eye and ear. Optogenetic stimulation promises to overcome fundamental shortcomings of electrical stimulation, namely poor spatial resolution and cellular specificity, and accordingly to deliver more detailed sensory information. This review aims at providing a comprehensive reference on current gene therapeutic and optogenetic research relevant to the restoration of hearing and vision. We will introduce gene-therapeutic approaches and discuss the biotechnological and optoelectronic aspects of optogenetic hearing and vision restoration.
Collapse
Affiliation(s)
| | | | | | | | - Tobias Moser
- Institute for Auditory Neuroscience, University Medical Center Goettingen, Germany
| |
Collapse
|
14
|
Schacker M, Seimetz D. From fiction to science: clinical potentials and regulatory considerations of gene editing. Clin Transl Med 2019; 8:27. [PMID: 31637541 PMCID: PMC6803602 DOI: 10.1186/s40169-019-0244-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
Gene editing technologies such as CRISPR/Cas9 have emerged as an attractive tool not only for scientific research but also for the development of medicinal products. Their ability to induce precise double strand breaks into DNA enables targeted modifications of the genome including selective knockout of genes, correction of mutations or precise insertion of new genetic material into specific loci. Gene editing-based therapies hold a great potential for the treatment of numerous diseases and the first products are already being tested in clinical trials. The treatment indications include oncological malignancies, HIV, diseases of the hematopoietic system and metabolic disorders. This article reviews ongoing preclinical and clinical studies and discusses how gene editing technologies are altering the gene therapy landscape. In addition, it focusses on the regulatory challenges associated with such therapies and how they can be tackled during the drug development process.
Collapse
Affiliation(s)
- Maria Schacker
- Biopharma Excellence GmbH, Agnes-Pockels-Bogen 1, 80992, Munich, Germany.
| | - Diane Seimetz
- Biopharma Excellence GmbH, Agnes-Pockels-Bogen 1, 80992, Munich, Germany
| |
Collapse
|
15
|
Chen T, Cao F, Wu X, Peng W, Zhi W, Weng J, Zhang X, Wang J. Constructing Gene-Enhanced Tissue Engineering for Regeneration and Repair of Osteochondral Defects. ACTA ACUST UNITED AC 2019; 3:e1900004. [PMID: 32648702 DOI: 10.1002/adbi.201900004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 06/06/2019] [Indexed: 11/07/2022]
Abstract
In situ sustained release of endogenous growth factors from cells is a challenge for repair and regeneration of tissue. Although recombinant adenovirus vectors are an effective delivery system that can prolong the release of growth factors and is very suitable for the therapy of growth factors, these recombinant adenovirus vectors that are widely used at present have low safety and stability in terms of long-term expression. In this study, the above problems are solved by knocking out both E1 and E3 genes at the same time and directly inserting the gene fragments encoding target proteins after the inverted terminal repeats. Finally, the combination of gene therapy with tissue engineering in regeneration and repair of full-thickness defects of osteochondral tissue are applied as an example. The results show that this strategy can achieve complete repair of articular osteochondral defects and recovery of their function, and meanwhile solve the problems of low safety and expression instability of recombinant adenovirus vectors. This method provides a bright prospect for the application of gene enhanced tissue engineering in the regeneration and repair of joint tissue, and also provides a reference for the repair and regeneration of other tissues.
Collapse
Affiliation(s)
- Taijun Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Fei Cao
- Department of Orthopaedics, Chengdu First People's Hospital, Chengdu, 610041, P. R. China
| | - Xin Wu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Wenzhen Peng
- Department of Biochemistry and Molecular Biology, College of Basic and Forensic Medicine, Sichuan University, Chengdu, 610041, P. R. China
| | - Wei Zhi
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Jie Weng
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xingdong Zhang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- National Engineering Research Center for Biomaterials, Engineering Research Center in Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Jianxin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
16
|
Goswami R, Subramanian G, Silayeva L, Newkirk I, Doctor D, Chawla K, Chattopadhyay S, Chandra D, Chilukuri N, Betapudi V. Gene Therapy Leaves a Vicious Cycle. Front Oncol 2019; 9:297. [PMID: 31069169 PMCID: PMC6491712 DOI: 10.3389/fonc.2019.00297] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022] Open
Abstract
The human genetic code encrypted in thousands of genes holds the secret for synthesis of proteins that drive all biological processes necessary for normal life and death. Though the genetic ciphering remains unchanged through generations, some genes get disrupted, deleted and or mutated, manifesting diseases, and or disorders. Current treatment options—chemotherapy, protein therapy, radiotherapy, and surgery available for no more than 500 diseases—neither cure nor prevent genetic errors but often cause many side effects. However, gene therapy, colloquially called “living drug,” provides a one-time treatment option by rewriting or fixing errors in the natural genetic ciphering. Since gene therapy is predominantly a viral vector-based medicine, it has met with a fair bit of skepticism from both the science fraternity and patients. Now, thanks to advancements in gene editing and recombinant viral vector development, the interest of clinicians and pharmaceutical industries has been rekindled. With the advent of more than 12 different gene therapy drugs for curing cancer, blindness, immune, and neuronal disorders, this emerging experimental medicine has yet again come in the limelight. The present review article delves into the popular viral vectors used in gene therapy, advances, challenges, and perspectives.
Collapse
Affiliation(s)
- Reena Goswami
- Neuroscience Branch, Research Division, United States Army Medical Research Institute of Chemical Defense, Aberdeen, MD, United States
| | - Gayatri Subramanian
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Liliya Silayeva
- Neuroscience Branch, Research Division, United States Army Medical Research Institute of Chemical Defense, Aberdeen, MD, United States
| | - Isabelle Newkirk
- Neuroscience Branch, Research Division, United States Army Medical Research Institute of Chemical Defense, Aberdeen, MD, United States
| | - Deborah Doctor
- Neuroscience Branch, Research Division, United States Army Medical Research Institute of Chemical Defense, Aberdeen, MD, United States
| | - Karan Chawla
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Saurabh Chattopadhyay
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Dhyan Chandra
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Nageswararao Chilukuri
- Neuroscience Branch, Research Division, United States Army Medical Research Institute of Chemical Defense, Aberdeen, MD, United States
| | - Venkaiah Betapudi
- Neuroscience Branch, Research Division, United States Army Medical Research Institute of Chemical Defense, Aberdeen, MD, United States.,Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
17
|
Osada T, Hartman ZC, Wei J, Lei G, Hobeika AC, Gwin WR, Diniz MA, Spector N, Clay TM, Chen W, Morse MA, Lyerly HK. Polyfunctional anti-human epidermal growth factor receptor 3 (anti-HER3) antibodies induced by HER3 vaccines have multiple mechanisms of antitumor activity against therapy resistant and triple negative breast cancers. Breast Cancer Res 2018; 20:90. [PMID: 30092835 PMCID: PMC6085609 DOI: 10.1186/s13058-018-1023-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 07/18/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Upregulation of human epidermal growth factor receptor 3 (HER3) is a major mechanism of acquired resistance to therapies targeting its heterodimerization partners epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2), but also exposes HER3 as a target for immune attack. We generated an adenovirus encoding full length human HER3 (Ad-HER3) to serve as a cancer vaccine. Previously we reported the anti-tumor efficacy and function of the T cell response to this vaccine. We now provide a detailed assessment of the antitumor efficacy and functional mechanisms of the HER3 vaccine-induced antibodies (HER3-VIAs) in serum from mice immunized with Ad-HER3. METHODS Serum containing HER3-VIA was tested in complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) assays and for its effect on HER3 internalization and degradation, downstream signaling of HER3 heterodimers and growth of metastatic HER2+ (BT474M1), HER2 therapy-resistant (rBT474), and triple negative (MDA-MB-468) breast cancers. RESULTS HER3-VIAs mediated CDC and ADCC, HER3 internalization, interruption of HER3 heterodimer-driven tumor signaling pathways, and anti-proliferative effects against HER2+ tumor cells in vitro and significant antitumor effects against metastatic HER2+ BT474M1, treatment refractory HER2+ rBT474 and triple negative MDA-MB-468 in vivo. CONCLUSIONS In addition to the T cell anti-tumor response induced by Ad-HER3, the HER3-VIAs provide additional functions to eliminate tumors in which HER3 signaling mediates aggressive behavior or acquired resistance to HER2-targeted therapy. These data support clinical studies of vaccination against HER3 prior to or concomitantly with other therapies to prevent outgrowth of therapy-resistant HER2+ and triple negative clones.
Collapse
Affiliation(s)
- Takuya Osada
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, MSRB Research Drive, Box 2714, Durham, NC, 27710, USA
| | - Zachary C Hartman
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, MSRB Research Drive, Box 2714, Durham, NC, 27710, USA
| | - Junping Wei
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, MSRB Research Drive, Box 2714, Durham, NC, 27710, USA
| | - Gangjun Lei
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, MSRB Research Drive, Box 2714, Durham, NC, 27710, USA
| | - Amy C Hobeika
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, MSRB Research Drive, Box 2714, Durham, NC, 27710, USA
| | - William R Gwin
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Marcio A Diniz
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Neil Spector
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Timothy M Clay
- Cell and Gene Therapy Discovery Research, PTS, GlaxoSmithKline, Collegeville, PA, USA
- Division of General Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Wei Chen
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Michael A Morse
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - H Kim Lyerly
- Division of Surgical Sciences, Department of Surgery, Duke University Medical Center, MSRB Research Drive, Box 2714, Durham, NC, 27710, USA.
| |
Collapse
|
18
|
Nattress CB, Halldén G. Advances in oncolytic adenovirus therapy for pancreatic cancer. Cancer Lett 2018; 434:56-69. [PMID: 29981812 DOI: 10.1016/j.canlet.2018.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023]
Abstract
Survival rates for pancreatic cancer patients have remained unchanged for the last four decades. The most aggressive, and most common, type of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC), which has the lowest 5-year survival rate of all cancers globally. The poor prognosis is typically due to late presentation of often non-specific symptoms and rapid development of resistance to all current therapeutics, including the standard-of-care cytotoxic drug gemcitabine. While early surgical intervention can significantly prolong patient survival, there are few treatment options for late-stage non-resectable metastatic disease, resulting in mostly palliative care. In addition, a defining feature of pancreatic cancer is the immunosuppressive and impenetrable desmoplastic stroma that blocks access to tumour cells by therapeutic drugs. The limited effectiveness of conventional chemotherapeutics reveals an urgent need to develop novel therapies with different mechanisms of action for this malignancy. An emerging alternative to current therapeutics is oncolytic adenoviruses; these engineered biological agents have proven efficacy and tumour-selectivity in preclinical pancreatic cancer models, including models of drug-resistant cancer. Safety of oncolytic adenoviral mutants has been extensively assessed in clinical trials with only limited toxicity to normal healthy tissue being reported. Promising efficacy in combination with gemcitabine was demonstrated in preclinical and clinical studies. A recent surge in novel adenoviral mutants entering clinical trials for pancreatic cancer indicates improved efficacy through activation of the host anti-tumour responses. The potential for adenoviruses to synergise with chemotherapeutics, activate anti-tumour immune responses, and contribute to stromal dissemination render these mutants highly attractive candidates for improved patient outcomes. Currently, momentum is gathering towards the development of systemically-deliverable mutants that are able to overcome anti-viral host immune responses, erythrocyte binding and hepatic uptake, to promote elimination of primary and metastatic lesions. This review will cover the key components of pancreatic cancer oncogenesis; novel oncolytic adenoviruses; clinical trials; and the current progress in overcoming the challenges of systemic delivery.
Collapse
Affiliation(s)
- Callum Baird Nattress
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, United Kingdom
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, United Kingdom.
| |
Collapse
|
19
|
Sharon D, Kamen A. Advancements in the design and scalable production of viral gene transfer vectors. Biotechnol Bioeng 2017; 115:25-40. [PMID: 28941274 DOI: 10.1002/bit.26461] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/16/2017] [Accepted: 09/19/2017] [Indexed: 01/22/2023]
Abstract
The last 10 years have seen a rapid expansion in the use of viral gene transfer vectors, with approved therapies and late stage clinical trials underway for the treatment of genetic disorders, and multiple forms of cancer, as well as prevention of infectious diseases through vaccination. With this increased interest and widespread adoption of viral vectors by clinicians and biopharmaceutical industries, there is an imperative to engineer safer and more efficacious vectors, and develop robust, scalable and cost-effective production platforms for industrialization. This review will focus on major innovations in viral vector design and production systems for three of the most widely used viral vectors: Adenovirus, Adeno-Associated Virus, and Lentivirus.
Collapse
Affiliation(s)
- David Sharon
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Amine Kamen
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
20
|
Decreased Vector Gene Expression from E2b Gene-Deleted Adenovirus Serotype 5 Vaccines Intensifies Proinflammatory Immune Responses. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00061-17. [PMID: 28381403 DOI: 10.1128/cvi.00061-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 03/20/2017] [Indexed: 02/06/2023]
Abstract
Recombinant adenovirus serotype 5 (Ad5) vectors are promising vaccine candidates due to their intrinsic immunogenicity and potent transgene expression; however, widespread preexisting Ad5 immunity has been considered a developmental impediment to the use of traditional, or conventional, E1 and E3 gene-deleted Ad5 (Ad5[E1-]) vaccines. Even in the presence of anti-Ad5 immunity, recent murine and human studies have confirmed E2b gene-deleted Ad5 (Ad5[E1-,E2b-]) vaccines to be highly efficacious inducers of transgene-specific memory responses and significantly less toxic options than Ad5[E1-] vaccines. While these findings have been substantially confirmed, the molecular mechanisms underlying the different reactions to these vaccine platforms are unknown. Using cultures of human peripheral blood mononuclear cells (hPBMCs) derived from multiple human donors, we found that Ad5[E1-,E2b-] vaccines trigger higher levels of hPBMC proinflammatory cytokine secretion than Ad5[E1-] vaccines. Interestingly, these responses were generated regardless of the donors' preexisting anti-Ad5 humoral and cell-mediated immune response status. In vitro hPBMC infection with the Ad5[E1-,E2b-] vaccine also provoked greater Th1-dominant gene responses yet smaller amounts of Ad-derived gene expression than Ad5[E1-] vaccines. These results suggest that Ad5[E1-,E2b-] vaccines, in contrast to Ad5[E1-] vaccines, do not promote activities that suppress innate immune signaling, thereby allowing for improved vaccine efficacy and a superior safety profile independently of previous Ad5 immunity.
Collapse
|
21
|
Sakhawat A, Liu Y, Ma L, Muhammad T, Wang S, Zhang L, Cong X, Huang Y. Upregulation of Coxsackie Adenovirus Receptor Sensitizes Cisplatin-Resistant Lung Cancer Cells to CRAd-Induced Inhibition. J Cancer 2017. [PMID: 28638457 PMCID: PMC5479248 DOI: 10.7150/jca.18371] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Objective. Conditionally replicating adenoviruses (CRAds) have been proven potent oncolytic viruses in previous studies. They selectively replicate in the tumor cells because of incorporated survivin promoter and ultimately lead to their killing with minimal side effects on normal tissue. Chemotherapy with cisplatin is commonly employed for treating tumors, but its cytotoxic effects and development of resistance remained major concerns to be dealt with. The aim of this study was to explore the anticancer potential of survivin regulated CRAd alone or in combination with cisplatin in the A549 lung cancer cell line and cisplatin-resistant lung cancer cell line, A549-DDPR. Methods. CRAd was genetically engineered in our laboratory by removing its E1B region and adding survivin promoter to control its replication. A549, H292, and H661 lung cancer cell lines were procured from the CAS-China. The anti-tumor effectiveness of combined treatment (cisplatin plus CRAd) was evaluated in vitro through MTS assays and in vivo through mouse model experimentation. RT- PCR was used to assess MDR gene and mRNA expression of coxsackie adenoviral receptor (CAR). Results. Results of in vitro studies established that A549 lung cancer cells were highly sensitive to cisplatin showing dose-dependent inhibition. The resistant cells of A549-DDPR exhibited very less sensitivity to cisplatin but were infected with CRAd more efficiently as compared to A549. A549-DDPR cells exhibited higher expression of MDR gene and CAR in the RT-PCR analysis. The nearly similar rise in the CAR expression was seen when lung cancer cell lines received cisplatin in combined treatment (cisplatin plus CRAd). Combined anti-cancer therapy (cisplatin plus oncolytic virus) proved more efficient than monotherapy in the killing of cancer cells. Results of in vivo experiments recapitulated nearly similar tumor inhibition activities. Conclusion. This study highlighted the significant role of survivin in gene therapy as it has the potential to render CRAd more tumor specific. It also establishes that higher CAR expression plays a vital role in the success of adenovirus-based therapies. Furthermore, a careful combination of chemotherapy drugs and oncolytic viruses can culminate in significant therapeutic achievements against cancer.
Collapse
Affiliation(s)
- Ali Sakhawat
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, China
| | - Yanan Liu
- Basic Medical College, Jilin University, China
| | - Ling Ma
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, China
| | - Tahir Muhammad
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, China
| | - Shensen Wang
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, China
| | - Lina Zhang
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, China
| | | | - Yinghui Huang
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, China
| |
Collapse
|
22
|
Afkhami S, LeClair DA, Haddadi S, Lai R, Toniolo SP, Ertl HC, Cranston ED, Thompson MR, Xing Z. Spray dried human and chimpanzee adenoviral-vectored vaccines are thermally stable and immunogenic in vivo. Vaccine 2017; 35:2916-2924. [PMID: 28438408 DOI: 10.1016/j.vaccine.2017.04.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 11/28/2022]
Abstract
Cold chain-free vaccine technologies are needed to ensure effective vaccine delivery and coverage, particularly in resource-poor countries. However, the immunogenicity and thermostability of spray dried live viral vector-based vaccines such as recombinant adenoviral-vectored vaccines remain to be investigated. To address this issue, we have spray dried human adenoviral (AdHu5)- and chimpanzee adenoviral (AdCh68)-vectored tuberculosis vaccines in a mannitol and dextran matrix. Spray dried powders containing these two vaccines display the morphologic and chemical properties desired for long-term thermostability and vaccination. Upon reconstitution, they effectively transfected the cells in vitro with relatively small losses in viral infectivity related to the spray drying process. Following in vivo vaccination, AdHu5- and AdCh68-vectored vaccines were as immunogenic as the conventional fresh, cryopreserved liquid vaccine samples. Of importance, even after cold chain-free storage, at ambient temperatures and relatively low humidity for 30 and 90days, the vaccines retained their in vivo immunogenicity, while the liquid vaccine samples stored under the same conditions lost their immune-activating capability almost entirely. Our results support further development of our spray drying technologies for generating thermally stable adenoviral-vectored and other viral-vectored vaccines.
Collapse
Affiliation(s)
- Sam Afkhami
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Daniel A LeClair
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Siamak Haddadi
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rocky Lai
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Steven P Toniolo
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Hildegund C Ertl
- Department of Immunology, The Wistar Institute, Philadelphia, PA, USA
| | - Emily D Cranston
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Michael R Thompson
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada.
| | - Zhou Xing
- McMaster Immunology Research Centre and Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
23
|
Osada T, Morse MA, Hobeika A, Diniz MA, Gwin WR, Hartman Z, Wei J, Guo H, Yang XY, Liu CX, Kaneko K, Broadwater G, Lyerly HK. Vaccination targeting human HER3 alters the phenotype of infiltrating T cells and responses to immune checkpoint inhibition. Oncoimmunology 2017; 6:e1315495. [PMID: 28680745 DOI: 10.1080/2162402x.2017.1315495] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023] Open
Abstract
Expression of human epidermal growth factor family member 3 (HER3), a critical heterodimerization partner with EGFR and HER2, promotes more aggressive biology in breast and other epithelial malignancies. As such, inhibiting HER3 could have broad applicability to the treatment of EGFR- and HER2-driven tumors. Although lack of a functional kinase domain limits the use of receptor tyrosine kinase inhibitors, HER3 contains antigenic targets for T cells and antibodies. Using novel human HER3 transgenic mouse models of breast cancer, we demonstrate that immunization with recombinant adenoviral vectors encoding full length human HER3 (Ad-HER3-FL) induces HER3-specific T cells and antibodies, alters the T cell infiltrate in tumors, and influences responses to immune checkpoint inhibitions. Both preventative and therapeutic Ad-HER3-FL immunization delayed tumor growth but were associated with both intratumoral PD-1 expressing CD8+ T cells and regulatory CD4+ T cell infiltrates. Immune checkpoint inhibition with either anti-PD-1 or anti-PD-L1 antibodies increased intratumoral CD8+ T cell infiltration and eliminated tumor following preventive vaccination with Ad-HER3-FL vaccine. The combination of dual PD-1/PD-L1 and CTLA4 blockade slowed the growth of tumor in response to Ad-HER3-FL in the therapeutic model. We conclude that HER3-targeting vaccines activate HER3-specific T cells and induce anti-HER3 specific antibodies, which alters the intratumoral T cell infiltrate and responses to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Takuya Osada
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Michael A Morse
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Marcio A Diniz
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | - William R Gwin
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA, USA
| | - Zachary Hartman
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Junping Wei
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Hongtao Guo
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Xiao-Yi Yang
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Cong-Xiao Liu
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Kensuke Kaneko
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Gloria Broadwater
- Duke University, Division of Biostatistics Duke Cancer Institute, Durham, NC, USA
| | - H Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
24
|
Gabitzsch ES, Tsang KY, Palena C, David JM, Fantini M, Kwilas A, Rice AE, Latchman Y, Hodge JW, Gulley JL, Madan RA, Heery CR, Balint JP, Jones FR, Schlom J. The generation and analyses of a novel combination of recombinant adenovirus vaccines targeting three tumor antigens as an immunotherapeutic. Oncotarget 2016; 6:31344-59. [PMID: 26374823 PMCID: PMC4741610 DOI: 10.18632/oncotarget.5181] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/24/2015] [Indexed: 12/11/2022] Open
Abstract
Phenotypic heterogeneity of human carcinoma lesions, including heterogeneity in expression of tumor-associated antigens (TAAs), is a well-established phenomenon. Carcinoembryonic antigen (CEA), MUC1, and brachyury are diverse TAAs, each of which is expressed on a wide range of human tumors. We have previously reported on a novel adenovirus serotype 5 (Ad5) vector gene delivery platform (Ad5 [E1-, E2b-]) in which regions of the early 1 (E1), early 2 (E2b), and early 3 (E3) genes have been deleted. The unique deletions in this platform result in a dramatic decrease in late gene expression, leading to a marked reduction in host immune response to the vector. Ad5 [E1-, E2b-]-CEA vaccine (ETBX-011) has been employed in clinical studies as an active vaccine to induce immune responses to CEA in metastatic colorectal cancer patients. We report here the development of novel recombinant Ad5 [E1-, E2b-]-brachyury and-MUC1 vaccine constructs, each capable of activating antigen-specific human T cells in vitro and inducing antigen-specific CD4+ and CD8+ T cells in vaccinated mice. We also describe the use of a combination of the three vaccines (designated Tri-Ad5) of Ad5 [E1-, E2b-]-CEA, Ad5 [E1-, E2b-]-brachyury and Ad5 [E1-, E2b-]-MUC1, and demonstrate that there is minimal to no “antigenic competition” in in vitro studies of human dendritic cells, or in murine vaccination studies. The studies reported herein support the rationale for the application of Tri-Ad5 as a therapeutic modality to induce immune responses to a diverse range of human TAAs for potential clinical studies.
Collapse
Affiliation(s)
| | - Kwong Yok Tsang
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin M David
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Massimo Fantini
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anna Kwilas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
25
|
LeClair DA, Cranston ED, Xing Z, Thompson MR. Evaluation of excipients for enhanced thermal stabilization of a human type 5 adenoviral vector through spray drying. Int J Pharm 2016; 506:289-301. [PMID: 27130366 DOI: 10.1016/j.ijpharm.2016.04.067] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/10/2016] [Accepted: 04/25/2016] [Indexed: 02/07/2023]
Abstract
We have produced a thermally stable recombinant human type 5 adenoviral vector (AdHu5) through spray drying with three excipient formulations (l-leucine, lactose/trehalose and mannitol/dextran). Spray drying leads to immobilization of the viral vector which is believed to prevent viral protein unfolding, aggregation and inactivation. The spray dried powders were characterized by scanning electron microscopy, differential scanning calorimetry, Karl Fischer titrations, and X-ray diffraction to identify the effects of temperature and atmospheric moisture on the immobilizing matrix. Thermal stability of the viral vector was confirmed in vitro by infection of A549 lung epithelial cells. Mannitol/dextran powders showed the greatest improvement in thermal stability with almost no viral activity loss after storage at 20°C for 90days (0.7±0.3 log TCID50) which is a significant improvement over the current -80°C storage protocol. Furthermore, viral activity was retained over short term exposure (72h) to temperatures as high as 55°C. Conversely, all powders exhibited activity loss when subjected to moisture due to amplified molecular motion of the matrix. Overall, a straightforward method ideal for the production of thermally stable vaccines has been demonstrated through spray drying AdHu5 with a blend of mannitol and dextran and storing the powder under low humidity conditions.
Collapse
Affiliation(s)
- Daniel A LeClair
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L8S 4L7, Canada
| | - Emily D Cranston
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L8S 4L7, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre & Department of Pathology and Molecular Medicine, McMaster University, Canada
| | - Michael R Thompson
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L8S 4L7, Canada.
| |
Collapse
|
26
|
|
27
|
Rice AE, Latchman YE, Balint JP, Lee JH, Gabitzsch ES, Jones FR. An HPV-E6/E7 immunotherapy plus PD-1 checkpoint inhibition results in tumor regression and reduction in PD-L1 expression. Cancer Gene Ther 2015; 22:454-62. [DOI: 10.1038/cgt.2015.40] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/05/2015] [Indexed: 12/29/2022]
|
28
|
Balint JP, Gabitzsch ES, Rice A, Latchman Y, Xu Y, Messerschmidt GL, Chaudhry A, Morse MA, Jones FR. Extended evaluation of a phase 1/2 trial on dosing, safety, immunogenicity, and overall survival after immunizations with an advanced-generation Ad5 [E1-, E2b-]-CEA(6D) vaccine in late-stage colorectal cancer. Cancer Immunol Immunother 2015; 64:977-87. [PMID: 25956394 PMCID: PMC4506904 DOI: 10.1007/s00262-015-1706-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/25/2015] [Indexed: 01/20/2023]
Abstract
A phase 1/2 clinical trial evaluating dosing, safety, immunogenicity, and overall survival on metastatic colorectal cancer (mCRC) patients after immunotherapy with an advanced-generation Ad5 [E1-, E2b-]-CEA(6D) vaccine was performed. We report our extended observations on long-term overall survival and further immune analyses on a subset of treated patients including assessment of cytolytic T cell responses, T regulatory (Treg) to T effector (Teff) cell ratios, flow cytometry on peripheral blood mononuclear cells (PBMCs), and determination of HLA-A2 status. An overall survival of 20 % (median survival 11 months) was observed during long-term follow-up, and no long-term adverse effects were reported. Cytolytic T cell responses increased after immunizations, and cell-mediated immune (CMI) responses were induced whether or not patients were HLA-A2 positive or Ad5 immune. PBMC samples from a small subset of patients were available for follow-up immune analyses. It was observed that the levels of carcinoembryonic antigen (CEA)-specific CMI activity decreased from their peak values during follow-up in five patients analyzed. Preliminary results revealed that activated CD4+ and CD8+ T cells were detected in a post-immunization sample exhibiting high CMI activity. Treg to Teff cell ratios were assessed, and samples from three of five patients exhibited a decrease in Treg to Teff cell ratio during the treatment protocol. Based upon the favorable safety and immunogenicity data obtained, we plan to perform an extensive immunologic and survival analysis on mCRC patients to be enrolled in a randomized/controlled clinical trial that investigates Ad5 [E1-, E2b-]-CEA(6D) as a single agent with booster immunizations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Michael A. Morse
- Department of Medicine, Duke University Medical Center, Durham, NC
| | | |
Collapse
|
29
|
Serguera C, Bemelmans AP. Gene therapy of the central nervous system: general considerations on viral vectors for gene transfer into the brain. Rev Neurol (Paris) 2014; 170:727-38. [PMID: 25459120 DOI: 10.1016/j.neurol.2014.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/13/2014] [Accepted: 09/10/2014] [Indexed: 02/04/2023]
Abstract
The last decade has nourished strong doubts on the beneficial prospects of gene therapy for curing fatal diseases. However, this climate of reservation is currently being transcended by the publication of several successful clinical protocols, restoring confidence in the appropriateness of therapeutic gene transfer. A strong sign of this present enthusiasm for gene therapy by clinicians and industrials is the market approval of the therapeutic viral vector Glybera, the first commercial product in Europe of this class of drug. This new field of medicine is particularly attractive when considering therapies for a number of neurological disorders, most of which are desperately waiting for a satisfactory treatment. The central nervous system is indeed a very compliant organ where gene transfer can be stable and successful if provided through an appropriate strategy. The purpose of this review is to present the characteristics of the most efficient virus-derived vectors used by researchers and clinicians to genetically modify particular cell types or whole regions of the brain. In addition, we discuss major issues regarding side effects, such as genotoxicity and immune response associated to the use of these vectors.
Collapse
Affiliation(s)
- C Serguera
- CEA, DSV, I(2)BM, Molecular Imaging Research Center (MIRCen) and CNRS, CEA URA 2210, 18, route du Panorama, 92265 Fontenay-aux-Roses, France
| | - A-P Bemelmans
- CEA, DSV, I(2)BM, Molecular Imaging Research Center (MIRCen) and CNRS, CEA URA 2210, 18, route du Panorama, 92265 Fontenay-aux-Roses, France.
| |
Collapse
|
30
|
Koestler BJ, Seregin SS, Rastall DPW, Aldhamen YA, Godbehere S, Amalfitano A, Waters CM. Stimulation of innate immunity by in vivo cyclic di-GMP synthesis using adenovirus. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:1550-9. [PMID: 25230938 PMCID: PMC4248757 DOI: 10.1128/cvi.00471-14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/11/2014] [Indexed: 12/19/2022]
Abstract
The bacterial second messenger cyclic di-GMP (c-di-GMP) stimulates inflammation by initiating innate immune cell recruitment and triggering the release of proinflammatory cytokines and chemokines. These properties make c-di-GMP a promising candidate for use as a vaccine adjuvant, and numerous studies have demonstrated that administration of purified c-di-GMP with different antigens increases protection against infection in animal models. Here, we have developed a novel approach to produce c-di-GMP inside host cells as an adjuvant to exploit a host-pathogen interaction and initiate an innate immune response. We have demonstrated that c-di-GMP can be synthesized in vivo by transducing a diguanylate cyclase (DGC) gene into mammalian cells using an adenovirus serotype 5 (Ad5) vector. Expression of DGC led to the production of c-di-GMP in vitro and in vivo, and this was able to alter proinflammatory gene expression in murine tissues and increase the secretion of numerous cytokines and chemokines when administered to animals. Furthermore, coexpression of DGC modestly increased T-cell responses to a Clostridium difficile antigen expressed from an adenovirus vaccine, although no significant differences in antibody titers were observed. This adenovirus c-di-GMP delivery system offers a novel method to administer c-di-GMP as an adjuvant to stimulate innate immunity during vaccination.
Collapse
Affiliation(s)
- Benjamin J Koestler
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Sergey S Seregin
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - David P W Rastall
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Sarah Godbehere
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| | - Christopher M Waters
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA The BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
31
|
|
32
|
Pan B, Géléoc GS, Asai Y, Horwitz GC, Kurima K, Ishikawa K, Kawashima Y, Griffith AJ, Holt JR. TMC1 and TMC2 are components of the mechanotransduction channel in hair cells of the mammalian inner ear. Neuron 2013; 79:504-15. [PMID: 23871232 DOI: 10.1016/j.neuron.2013.06.019] [Citation(s) in RCA: 317] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2013] [Indexed: 02/02/2023]
Abstract
Sensory transduction in auditory and vestibular hair cells requires expression of transmembrane channel-like (Tmc) 1 and 2 genes, but the function of these genes is unknown. To investigate the hypothesis that TMC1 and TMC2 proteins are components of the mechanosensitive ion channels that convert mechanical information into electrical signals, we recorded whole-cell and single-channel currents from mouse hair cells that expressed Tmc1, Tmc2, or mutant Tmc1. Cells that expressed Tmc2 had high calcium permeability and large single-channel currents, while cells with mutant Tmc1 had reduced calcium permeability and reduced single-channel currents. Cells that expressed Tmc1 and Tmc2 had a broad range of single-channel currents, suggesting multiple heteromeric assemblies of TMC subunits. The data demonstrate TMC1 and TMC2 are components of hair cell transduction channels and contribute to permeation properties. Gradients in TMC channel composition may also contribute to variation in sensory transduction along the tonotopic axis of the mammalian cochlea.
Collapse
Affiliation(s)
- Bifeng Pan
- Department of Otolaryngology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Center for Life Sciences 12251, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ibrišimović M, Lion T, Klein R. Combinatorial targeting of 2 different steps in adenoviral DNA replication by herpes simplex virus thymidine kinase and artificial microRNA expression for the inhibition of virus multiplication in the presence of ganciclovir. BMC Biotechnol 2013; 13:54. [PMID: 23822768 PMCID: PMC3720212 DOI: 10.1186/1472-6750-13-54] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/19/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human adenoviruses are a frequent threat to immunocompromised patients, and disseminated disease is associated with severe morbidity and mortality. Current drugs are not capable of preventing all fatalities, thus indicating the need for alternative treatment strategies. Adenoviruses can be rendered susceptible to antiherpetic prodrugs such as ganciclovir (GCV), upon expression of the herpes simplex virus thymidine kinase (HSV-TK) gene in adenovirus-infected cells. Furthermore, adenoviruses are amenable to post-transcriptional gene silencing via small interfering RNAs (siRNAs) or artificial micro RNAs (amiRNAs). RESULTS In this study, we combined these 2 approaches by constructing a combinatorial gene expression cassette that comprises the HSV-TK gene and multiple copies of an amiRNA directed against the mRNA encoding the adenoviral preterminal protein (pTP). HSV-TK gene expression was controlled by the adenoviral E4 promoter, which is activated in the presence of the adenoviral E1 gene products (i.e., when adenovirus is present in the cell). When inserted into a replication-deficient (E1-, E3-deleted) adenoviral vector, this cassette effectively inhibited the replication of wild-type adenovirus in vitro. The reduction rate mediated by the combinatorial approach was higher compared to that achieved by either of the 2 approaches alone, and these obvious additive effects became most pronounced when the GCV concentration was low. CONCLUSIONS The concept presented here has the potential to aid in the inhibition of wild-type adenovirus replication. Furthermore, the combinatorial expression cassette may constitute a safeguard to potentially control unintended replication of adenoviral vectors and to prevent immune responses provoked by them.
Collapse
Affiliation(s)
- Mirza Ibrišimović
- Children's Cancer Research Institute, St, Anna Kinderkrebsforschung, Zimmermannplatz 10, 1090 Vienna, Austria
| | | | | |
Collapse
|
34
|
Morse MA, Chaudhry A, Gabitzsch ES, Hobeika AC, Osada T, Clay TM, Amalfitano A, Burnett BK, Devi GR, Hsu DS, Xu Y, Balcaitis S, Dua R, Nguyen S, Balint JP, Jones FR, Lyerly HK. Novel adenoviral vector induces T-cell responses despite anti-adenoviral neutralizing antibodies in colorectal cancer patients. Cancer Immunol Immunother 2013; 62:1293-301. [PMID: 23624851 DOI: 10.1007/s00262-013-1400-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 01/21/2013] [Indexed: 12/11/2022]
Abstract
First-generation, E1-deleted adenovirus subtype 5 (Ad5)-based vectors, although promising platforms for use as cancer vaccines, are impeded in activity by naturally occurring or induced Ad-specific neutralizing antibodies. Ad5-based vectors with deletions of the E1 and the E2b regions (Ad5 [E1-, E2b-]), the latter encoding the DNA polymerase and the pre-terminal protein, by virtue of diminished late phase viral protein expression, were hypothesized to avoid immunological clearance and induce more potent immune responses against the encoded tumor antigen transgene in Ad-immune hosts. Indeed, multiple homologous immunizations with Ad5 [E1-, E2b-]-CEA(6D), encoding the tumor antigen carcinoembryonic antigen (CEA), induced CEA-specific cell-mediated immune (CMI) responses with antitumor activity in mice despite the presence of preexisting or induced Ad5-neutralizing antibody. In the present phase I/II study, cohorts of patients with advanced colorectal cancer were immunized with escalating doses of Ad5 [E1-, E2b-]-CEA(6D). CEA-specific CMI responses were observed despite the presence of preexisting Ad5 immunity in a majority (61.3 %) of patients. Importantly, there was minimal toxicity, and overall patient survival (48 % at 12 months) was similar regardless of preexisting Ad5 neutralizing antibody titers. The results demonstrate that, in cancer patients, the novel Ad5 [E1-, E2b-] gene delivery platform generates significant CMI responses to the tumor antigen CEA in the setting of both naturally acquired and immunization-induced Ad5-specific immunity.
Collapse
Affiliation(s)
- Michael A Morse
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gan Y, Jing Z, Stetler RA, Cao G. Gene delivery with viral vectors for cerebrovascular diseases. Front Biosci (Elite Ed) 2013; 5:188-203. [PMID: 23276981 PMCID: PMC5516729 DOI: 10.2741/e607] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Recent achievements in the understanding of molecular events involved in the pathogenesis of central nervous system (CNS) injury have made gene transfer a promising approach for various neurological disorders, including cerebrovascular diseases. However, special obstacles, including the post-mitotic nature of neurons and the blood-brain barrier (BBB), constitute key challenges for gene delivery to the CNS. Despite the various limitations in current gene delivery systems, a spectrum of viral vectors has been successfully used to deliver genes to the CNS. Furthermore, recent advancements in vector engineering have improved the safety and delivery of viral vectors. Numerous viral vector-based clinical trials for neurological disorders have been initiated. This review will summarize the current implementation of viral gene delivery in the context of cerebrovascular diseases including ischemic stroke, hemorrhagic stroke and subarachnoid hemorrhage (SAH). In particular, we will discuss the potentially feasible ways in which viral vectors can be manipulated and exploited for use in neural delivery and therapy.
Collapse
Affiliation(s)
- Yu Gan
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, U.S.A
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| | - Zheng Jing
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, U.S.A
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| | - R. Anne Stetler
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, U.S.A
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| | - Guodong Cao
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, U.S.A
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| |
Collapse
|
36
|
Atkinson PJ, Wise AK, Flynn BO, Nayagam BA, Hume CR, O’Leary SJ, Shepherd RK, Richardson RT. Neurotrophin gene therapy for sustained neural preservation after deafness. PLoS One 2012; 7:e52338. [PMID: 23284995 PMCID: PMC3524079 DOI: 10.1371/journal.pone.0052338] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 11/14/2012] [Indexed: 11/18/2022] Open
Abstract
The cochlear implant provides auditory cues to profoundly deaf patients by electrically stimulating the residual spiral ganglion neurons. These neurons, however, undergo progressive degeneration after hearing loss, marked initially by peripheral fibre retraction and ultimately culminating in cell death. This research aims to use gene therapy techniques to both hold and reverse this degeneration by providing a sustained and localised source of neurotrophins to the deafened cochlea. Adenoviral vectors containing green fluorescent protein, with or without neurotrophin-3 and brain derived neurotrophic factor, were injected into the lower basal turn of scala media of guinea pigs ototoxically deafened one week prior to intervention. This single injection resulted in localised and sustained gene expression, principally in the supporting cells within the organ of Corti. Guinea pigs treated with adenoviral neurotrophin-gene therapy had greater neuronal survival compared to contralateral non-treated cochleae when examined at 7 and 11 weeks post injection. Moreover; there was evidence of directed peripheral fibre regrowth towards cells expressing neurotrophin genes after both treatment periods. These data suggest that neurotrophin-gene therapy can provide sustained protection of spiral ganglion neurons and peripheral fibres after hearing loss.
Collapse
Affiliation(s)
- Patrick J. Atkinson
- Bionics Institute, East Melbourne, Victoria, Australia
- Department of Otolaryngology, University of Melbourne, East Melbourne, Victoria, Australia
| | - Andrew K. Wise
- Bionics Institute, East Melbourne, Victoria, Australia
- Department of Otolaryngology, University of Melbourne, East Melbourne, Victoria, Australia
- Department of Medical Bionics, University of Melbourne, East Melbourne, Victoria, Australia
| | | | - Bryony A. Nayagam
- Bionics Institute, East Melbourne, Victoria, Australia
- Department of Otolaryngology, University of Melbourne, East Melbourne, Victoria, Australia
| | - Clifford R. Hume
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, Washington, United States of America
| | - Stephen J. O’Leary
- Bionics Institute, East Melbourne, Victoria, Australia
- Department of Otolaryngology, University of Melbourne, East Melbourne, Victoria, Australia
| | - Robert K. Shepherd
- Bionics Institute, East Melbourne, Victoria, Australia
- Department of Otolaryngology, University of Melbourne, East Melbourne, Victoria, Australia
- Department of Medical Bionics, University of Melbourne, East Melbourne, Victoria, Australia
| | - Rachael T. Richardson
- Bionics Institute, East Melbourne, Victoria, Australia
- Department of Otolaryngology, University of Melbourne, East Melbourne, Victoria, Australia
- Department of Medical Bionics, University of Melbourne, East Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
37
|
Lentz TB, Gray SJ, Samulski RJ. Viral vectors for gene delivery to the central nervous system. Neurobiol Dis 2012; 48:179-88. [PMID: 22001604 PMCID: PMC3293995 DOI: 10.1016/j.nbd.2011.09.014] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/17/2011] [Accepted: 09/29/2011] [Indexed: 12/19/2022] Open
Abstract
The potential benefits of gene therapy for neurological diseases such as Parkinson's, Amyotrophic Lateral Sclerosis (ALS), Epilepsy, and Alzheimer's are enormous. Even a delay in the onset of severe symptoms would be invaluable to patients suffering from these and other diseases. Significant effort has been placed in developing vectors capable of delivering therapeutic genes to the CNS in order to treat neurological disorders. At the forefront of potential vectors, viral systems have evolved to efficiently deliver their genetic material to a cell. The biology of different viruses offers unique solutions to the challenges of gene therapy, such as cell targeting, transgene expression and vector production. It is important to consider the natural biology of a vector when deciding whether it will be the most effective for a specific therapeutic function. In this review, we outline desired features of the ideal vector for gene delivery to the CNS and discuss how well available viral vectors compare to this model. Adeno-associated virus, retrovirus, adenovirus and herpesvirus vectors are covered. Focus is placed on features of the natural biology that have made these viruses effective tools for gene delivery with emphasis on their application in the CNS. Our goal is to provide insight into features of the optimal vector and which viral vectors can provide these features.
Collapse
Affiliation(s)
- Thomas B. Lentz
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steven J. Gray
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
38
|
Gabitzsch ES, Balint-Junior JP, Xu Y, Balcaitis S, Sanders-Beer B, Karl J, Weinhold KJ, Paessler S, Jones FR. Control of SIV infection and subsequent induction of pandemic H1N1 immunity in rhesus macaques using an Ad5 [E1-, E2b-] vector platform. Vaccine 2012; 30:7265-70. [PMID: 23041546 DOI: 10.1016/j.vaccine.2012.09.058] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 09/20/2012] [Accepted: 09/22/2012] [Indexed: 01/21/2023]
Abstract
Anti-vector immunity mitigates immune responses induced by recombinant adenovirus vector vaccines, limiting their prime-boost capabilities. We have developed a novel gene delivery and expression platform (Ad5 [E1-, E2b-]) that induces immune responses despite pre-existing and/or developed concomitant Ad5 immunity. In the present study, we evaluated if this new Ad5 platform could overcome the adverse condition of pre-existing Ad5 immunity to induce effective immune responses in prime-boost immunization regimens against two different infectious diseases in the same animal. Ad5 immune rhesus macaques (RM) were immunized multiple times with the Ad5 [E1-, E2b-] platform expressing antigens from simian immunodeficiency virus (SIV). Immunized RM developed cell-mediated immunity against SIV antigens Gag, Pol, Nef and Env as well as antibody against Env. Vaccinated and vector control RMs were challenged intra-rectally with homologous SIVmac239. During a 7-week follow-up, there was perturbation of SIV load in some immunized RM. At 7 weeks post-challenge, eight immunized animals (53%) did not have detectable SIV, compared to two RM controls (13%) (P<0.02; log-rank Mantel-Cox test). There was no correlation of protective MHC contributing to infection control. The RM without detectable circulating SIV, now hyper immune to Ad5, were then vaccinated with the same Ad5 [E1-, E2b-] platform expressing H1N1 influenza hemagglutinin (HA). Thirty days post Ad5 [E1-, E2b-]-HA vaccination, significant levels of influenza neutralizing antibody were induced in all animals that increased after an Ad5 [E1-, E2b-]-HA homologous boost. These data demonstrate the versatility of this new vector platform to immunize against two separate disease targets in the same animal despite the presence of immunity against the delivery platform, permitting homologous repeat immunizations with an Ad5 gene delivery platform.
Collapse
|
39
|
Wieking BG, Vermeer DW, Spanos WC, Lee KM, Vermeer P, Lee WT, Xu Y, Gabitzsch ES, Balcaitis S, Balint JP, Jones FR, Lee JH. A non-oncogenic HPV 16 E6/E7 vaccine enhances treatment of HPV expressing tumors. Cancer Gene Ther 2012; 19:667-74. [PMID: 22918471 PMCID: PMC3571619 DOI: 10.1038/cgt.2012.55] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Human papillomaviruses (HPVs) are the causative factor for >90% of cervical cancers and 25% of head and neck cancers. The incidence of HPV positive (+) head and neck squamous cell carcinomas has greatly increased in the last 30 years. E6 and E7 are the two key viral oncoproteins that induce and propagate cellular transformation. An immune response generated during cisplatin/radiation therapy improves tumor clearance of HPV(+) cancers. Augmenting this induced response during therapy with an adenoviral HPV16 E6/E7 vaccine improves long-term survival in pre-clinical models. Here, we describe the generation of an HPV16 E6/E7 construct, which contains mutations that render E6/E7 non-oncogenic, while preserving antigenicity. These mutations do not allow E6/E7 to degrade p53, pRb, PTPN13, or activate telomerase. Non-oncogenic E6/E7 (E6(Δ)/E7(Δ)) expressed as a stable integrant, or in the [E1-, E2b-] adenovirus, lacks the ability to transform human cells while retaining the ability to induce an HPV-specific immune response. Moreover, E6(Δ)/E7(Δ) plus chemotherapy/radiation statistically enhances clearance of established HPV(+) cancer in vivo.
Collapse
Affiliation(s)
- B G Wieking
- Cancer Biology Research Center, Sanford Research/University of South Dakota, Sioux Falls, SD 57104, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Adv Biomed Res 2012; 1:27. [PMID: 23210086 PMCID: PMC3507026 DOI: 10.4103/2277-9175.98152] [Citation(s) in RCA: 540] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 03/10/2012] [Indexed: 12/14/2022] Open
Abstract
Gene therapy is the process of introducing foreign genomic materials into host cells to elicit a therapeutic benefit. Although initially the main focus of gene therapy was on special genetic disorders, now diverse diseases with different patterns of inheritance and acquired diseases are targets of gene therapy. There are 2 major categories of gene therapy, including germline gene therapy and somatic gene therapy. Although germline gene therapy may have great potential, because it is currently ethically forbidden, it cannot be used; however, to date human gene therapy has been limited to somatic cells. Although numerous viral and nonviral gene delivery systems have been developed in the last 3 decades, no delivery system has been designed that can be applied in gene therapy of all kinds of cell types in vitro and in vivo with no limitation and side effects. In this review we explain about the history of gene therapy, all types of gene delivery systems for germline (nuclei, egg cells, embryonic stem cells, pronuclear, microinjection, sperm cells) and somatic cells by viral [retroviral, adenoviral, adeno association, helper-dependent adenoviral systems, hybrid adenoviral systems, herpes simplex, pox virus, lentivirus, Epstein-Barr virus)] and nonviral systems (physical: Naked DNA, DNA bombardant, electroporation, hydrodynamic, ultrasound, magnetofection) and (chemical: Cationic lipids, different cationic polymers, lipid polymers). In addition to the above-mentioned, advantages, disadvantages, and practical use of each system are discussed.
Collapse
Affiliation(s)
- Nouri Nayerossadat
- Molecular Genetic Laboratory, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Disease Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Talebi Maedeh
- Molecular Genetic Laboratory, Alzahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Palizban Abas Ali
- Department of Clinical Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences and Health Services, Isfahan, Iran
| |
Collapse
|
41
|
Sun H, Samarghandi A, Zhang N, Yao Z, Xiong M, Teng BB. Proprotein Convertase Subtilisin/Kexin Type 9 Interacts With Apolipoprotein B and Prevents Its Intracellular Degradation, Irrespective of the Low-Density Lipoprotein Receptor. Arterioscler Thromb Vasc Biol 2012; 32:1585-95. [DOI: 10.1161/atvbaha.112.250043] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
proprotein convertase subtilisin/kexin type 9 (PCSK9) negatively regulates the low-density lipoprotein (LDL) receptor (LDLR) in hepatocytes and therefore plays an important role in controlling circulating levels of LDL-cholesterol. To date, the relationship between PCSK9 and metabolism of apolipoprotein B (apoB), the structural protein of LDL, has been controversial and remains to be clarified.
Methods and Results—
We assessed the impact of PCSK9 overexpression (≈400-fold above baseline) on apoB synthesis and secretion in 3 mouse models: wild-type C57BL/6 mice and LDLR-null mice (
Ldlr
−/−
and
Ldlr
−/−
Apobec1
−/−
). Irrespective of LDLR expression, mice transduced with the
PCSK9
gene invariably exhibited increased levels of plasma cholesterol, triacylglycerol, and apoB. Consistent with these findings, the levels of very-low-density lipoprotein and LDL were also increased whereas high-density lipoprotein levels were unchanged. Importantly, we demonstrated that endogenous PCSK9 interacted with apoB in hepatocytes. The PCSK9/apoB interaction resulted in increased production of apoB, possibly through the inhibition of intracellular apoB degradation via the autophagosome/lysosome pathway.
Conclusion—
We propose a new role for PCSK9 that involves shuttling between apoB and LDLR. The present study thus provides new insights into the action of PCSK9 in regulating apoB metabolism. Furthermore, our results indicate that targeting PCSK9 expression represents a new paradigm in therapeutic intervention against hyperlipidemia.
Collapse
Affiliation(s)
- Hua Sun
- From the University of Texas Graduate School of Biomedical Sciences at Houston (H.S., B-B.T.); Center for Human Genetics (H.S., A.S., B-B.T.) and the Texas Therapeutics Institute (N.Z.), The Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, TX; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada (Z.Y.); and Human Genetics Center, School of Public Health, the University of Texas Health
| | - Amin Samarghandi
- From the University of Texas Graduate School of Biomedical Sciences at Houston (H.S., B-B.T.); Center for Human Genetics (H.S., A.S., B-B.T.) and the Texas Therapeutics Institute (N.Z.), The Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, TX; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada (Z.Y.); and Human Genetics Center, School of Public Health, the University of Texas Health
| | - Ningyan Zhang
- From the University of Texas Graduate School of Biomedical Sciences at Houston (H.S., B-B.T.); Center for Human Genetics (H.S., A.S., B-B.T.) and the Texas Therapeutics Institute (N.Z.), The Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, TX; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada (Z.Y.); and Human Genetics Center, School of Public Health, the University of Texas Health
| | - Zemin Yao
- From the University of Texas Graduate School of Biomedical Sciences at Houston (H.S., B-B.T.); Center for Human Genetics (H.S., A.S., B-B.T.) and the Texas Therapeutics Institute (N.Z.), The Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, TX; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada (Z.Y.); and Human Genetics Center, School of Public Health, the University of Texas Health
| | - Momiao Xiong
- From the University of Texas Graduate School of Biomedical Sciences at Houston (H.S., B-B.T.); Center for Human Genetics (H.S., A.S., B-B.T.) and the Texas Therapeutics Institute (N.Z.), The Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, TX; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada (Z.Y.); and Human Genetics Center, School of Public Health, the University of Texas Health
| | - Ba-Bie Teng
- From the University of Texas Graduate School of Biomedical Sciences at Houston (H.S., B-B.T.); Center for Human Genetics (H.S., A.S., B-B.T.) and the Texas Therapeutics Institute (N.Z.), The Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, TX; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada (Z.Y.); and Human Genetics Center, School of Public Health, the University of Texas Health
| |
Collapse
|
42
|
Malaria vaccines: focus on adenovirus based vectors. Vaccine 2012; 30:5191-8. [PMID: 22683663 DOI: 10.1016/j.vaccine.2012.05.048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/09/2012] [Accepted: 05/19/2012] [Indexed: 11/24/2022]
Abstract
Protection against malaria through vaccination is known to be achievable, as first demonstrated over 30 years ago. Vaccination via repeated bites with Plasmodium falciparum infected and irradiated mosquitoes provided short lived protection from malaria infection to these vaccinees. Though this method still remains the most protective malaria vaccine to date, it is likely impractical for widespread use. However, recent developments in sub-unit malaria vaccine platforms are bridging the gap between high levels of protection and feasibility. The current leading sub-unit vaccine, RTS,S (which consists of a fusion of a portion of the P. falciparum derived circumsporozoite protein to the Hepatitis B surface antigen), has demonstrated the ability to induce protection from malaria infection in up 56% of RTS,S vaccinees. Though encouraging, these results may fall short of protection levels generally considered to be required to achieve eradication of malaria. Therefore, the use of viral vectored vaccine platforms has recently been pursued to further improve the efficacy of malaria targeted vaccines. Adenovirus based vaccine platforms have demonstrated potent anti-malaria immune responses when used alone, as well when utilized in heterologous prime boost regimens. This review will provide an update as to the current advancements in malaria vaccine development, with a focus on the use of adenovirus vectored malaria vaccines.
Collapse
|
43
|
Levin ME, Holt JR. The function and molecular identity of inward rectifier channels in vestibular hair cells of the mouse inner ear. J Neurophysiol 2012; 108:175-86. [PMID: 22496522 DOI: 10.1152/jn.00098.2012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inner ear hair cells respond to mechanical stimuli with graded receptor potentials. These graded responses are modulated by a host of voltage-dependent currents that flow across the basolateral membrane. Here, we examine the molecular identity and the function of a class of voltage-dependent ion channels that carries the potassium-selective inward rectifier current known as I(K1). I(K1) has been identified in vestibular hair cells of various species, but its molecular composition and functional contributions remain obscure. We used quantitative RT-PCR to show that the inward rectifier gene, Kir2.1, is highly expressed in mouse utricle between embryonic day 15 and adulthood. We confirmed Kir2.1 protein expression in hair cells by immunolocalization. To examine the molecular composition of I(K1), we recorded voltage-dependent currents from type II hair cells in response to 50-ms steps from -124 to -54 in 10-mV increments. Wild-type cells had rapidly activating inward currents with reversal potentials close to the K(+) equilibrium potential and a whole-cell conductance of 4.8 ± 1.5 nS (n = 46). In utricle hair cells from Kir2.1-deficient (Kir2.1(-/-)) mice, I(K1) was absent at all stages examined. To identify the functional contribution of Kir2.1, we recorded membrane responses in current-clamp mode. Hair cells from Kir2.1(-/-) mice had significantly (P < 0.001) more depolarized resting potentials and larger, slower membrane responses than those of wild-type cells. These data suggest that Kir2.1 is required for I(K1) in type II utricle hair cells and contributes to hyperpolarized resting potentials and fast, small amplitude receptor potentials in response to current inputs, such as those evoked by hair bundle deflections.
Collapse
Affiliation(s)
- Michaela E Levin
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | |
Collapse
|
44
|
Abstract
Progress in vector design and an increased knowledge of mechanisms underlying tumor-induced immune suppression have led to a new and promising generation of Adenovirus (Ad)-based immunotherapies, which are discussed in this review. As vaccine vehicles Ad vectors (AdVs) have been clinically evaluated and proven safe, but a major limitation of the commonly used Ad5 serotype is neutralization by preexistent or rapidly induced immune responses. Genetic modifications in the Ad capsid can reduce intrinsic immunogenicity and facilitate escape from antibody-mediated neutralization. Further modification of the Ad hexon and fiber allows for liver and scavenger detargeting and selective targeting of, for example, dendritic cells. These next-generation Ad vaccines with enhanced efficacy are now becoming available for testing as tumor vaccines. In addition, AdVs encoding immune-modulating products may be used to convert the tumor microenvironment from immune-suppressive and proinvasive to proinflammatory, thus facilitating cell-mediated effector functions that can keep tumor growth and invasion in check. Oncolytic AdVs, that selectively replicate in tumor cells and induce an immunogenic form of cell death, can also be armed with immune-activating transgenes to amplify primed antitumor immune responses. These novel immunotherapy strategies, employing highly efficacious AdVs in optimized configurations, show great promise and warrant clinical exploration.
Collapse
|
45
|
Edry E, Lamprecht R, Wagner S, Rosenblum K. Virally mediated gene manipulation in the adult CNS. Front Mol Neurosci 2011; 4:57. [PMID: 22207836 PMCID: PMC3245970 DOI: 10.3389/fnmol.2011.00057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2011] [Accepted: 12/15/2011] [Indexed: 12/02/2022] Open
Abstract
Understanding how the CNS functions poses one of the greatest challenges in modern life science and medicine. Studying the brain is especially challenging because of its complexity, the heterogeneity of its cellular composition, and the substantial changes it undergoes throughout its life-span. The complexity of adult brain neural networks results also from the diversity of properties and functions of neuronal cells, governed, inter alia, by temporally and spatially differential expression of proteins in mammalian brain cell populations. Hence, research into the biology of CNS activity and its implications to human and animal behavior must use novel scientific tools. One source of such tools is the field of molecular genetics—recently utilized more and more frequently in neuroscience research. Transgenic approaches in general, and gene targeting in rodents have become fundamental tools for elucidating gene function in the CNS. Although spectacular progress has been achieved over recent decades by using these approaches, it is important to note that they face a number of restrictions. One of the main challenges is presented by the temporal and spatial regulation of introduced genetic manipulations. Viral vectors provide an alternative approach to temporally regulated, localized delivery of genetic modifications into neurons. In this review we describe available technologies for gene transfer into the adult mammalian CNS that use both viral and non-viral tools. We discuss viral vectors frequently used in neuroscience, with emphasis on lentiviral vector (LV) systems. We consider adverse effects of LVs, and the use of LVs for temporally and spatially controllable manipulations. Especially, we highlight the significance of viral vector-mediated genetic manipulations in studying learning and memory processes, and how they may be effectively used to separate out the various phases of learning: acquisition, consolidation, retrieval, and maintenance.
Collapse
Affiliation(s)
- Efrat Edry
- Department of Neurobiology and Ethology, Center for Gene Manipulation in the Brain (CGMB), University of Haifa Haifa, Israel
| | | | | | | |
Collapse
|
46
|
Kawashima Y, Géléoc GSG, Kurima K, Labay V, Lelli A, Asai Y, Makishima T, Wu DK, Della Santina CC, Holt JR, Griffith AJ. Mechanotransduction in mouse inner ear hair cells requires transmembrane channel-like genes. J Clin Invest 2011; 121:4796-809. [PMID: 22105175 DOI: 10.1172/jci60405] [Citation(s) in RCA: 320] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 10/19/2011] [Indexed: 01/22/2023] Open
Abstract
Inner ear hair cells convert the mechanical stimuli of sound, gravity, and head movement into electrical signals. This mechanotransduction process is initiated by opening of cation channels near the tips of hair cell stereocilia. Since the identity of these ion channels is unknown, and mutations in the gene encoding transmembrane channel-like 1 (TMC1) cause hearing loss without vestibular dysfunction in both mice and humans, we investigated the contribution of Tmc1 and the closely related Tmc2 to mechanotransduction in mice. We found that Tmc1 and Tmc2 were expressed in mouse vestibular and cochlear hair cells and that GFP-tagged TMC proteins localized near stereocilia tips. Tmc2 expression was transient in early postnatal mouse cochlear hair cells but persisted in vestibular hair cells. While mice with a targeted deletion of Tmc1 (Tmc1(Δ) mice) were deaf and those with a deletion of Tmc2 (Tmc2(Δ) mice) were phenotypically normal, Tmc1(Δ)Tmc2(Δ) mice had profound vestibular dysfunction, deafness, and structurally normal hair cells that lacked all mechanotransduction activity. Expression of either exogenous TMC1 or TMC2 rescued mechanotransduction in Tmc1(Δ)Tmc2(Δ) mutant hair cells. Our results indicate that TMC1 and TMC2 are necessary for hair cell mechanotransduction and may be integral components of the mechanotransduction complex. Our data also suggest that persistent TMC2 expression in vestibular hair cells may preserve vestibular function in humans with hearing loss caused by TMC1 mutations.
Collapse
Affiliation(s)
- Yoshiyuki Kawashima
- Molecular Biology and Genetics Section, National Institute on Deafness and Other Communication Disorders, NIH, Rockville, Maryland 20850-3320, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Aldhamen YA, Seregin SS, Amalfitano A. Immune recognition of gene transfer vectors: focus on adenovirus as a paradigm. Front Immunol 2011; 2:40. [PMID: 22566830 PMCID: PMC3342374 DOI: 10.3389/fimmu.2011.00040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 08/18/2011] [Indexed: 11/13/2022] Open
Abstract
Recombinant Adenovirus (Ad) based vectors have been utilized extensively as a gene transfer platform in multiple pre-clinical and clinical applications. These applications are numerous, and inclusive of both gene therapy and vaccine based approaches to human or animal diseases. The widespread utilization of these vectors in both animal models, as well as numerous human clinical trials (Ad-based vectors surpass all other gene transfer vectors relative to numbers of patients treated, as well as number of clinical trials overall), has shed light on how this virus vector interacts with both the innate and adaptive immune systems. The ability to generate and administer large amounts of this vector likely contributes not only to their ability to allow for highly efficient gene transfer, but also their elicitation of host immune responses to the vector and/or the transgene the vector expresses in vivo. These facts, coupled with utilization of several models that allow for full detection of these responses has predicted several observations made in human trials, an important point as lack of similar capabilities by other vector systems may prevent detection of such responses until only after human trials are initiated. Finally, induction of innate or adaptive immune responses by Ad vectors may be detrimental in one setting (i.e., gene therapy) and be entirely beneficial in another (i.e., prophylactic or therapeutic vaccine based applications). Herein, we review the current understanding of innate and adaptive immune responses to Ad vectors, as well some recent advances that attempt to capitalize on this understanding so as to further broaden the safe and efficient use of Ad-based gene transfer therapies in general.
Collapse
Affiliation(s)
- Yasser Ali Aldhamen
- Department of Microbiology and Molecular Genetics, Michigan State University East Lansing, MI, USA
| | | | | |
Collapse
|
48
|
Gabitzsch ES, Xu Y, Balint JP, Balcaitis S, Sanders-Beer B, Jones FR. Induction and comparison of SIV immunity in Ad5 naïve and Ad5 immune non-human primates using an Ad5 [E1-, E2b-] based vaccine. Vaccine 2011; 29:8101-7. [PMID: 21864618 DOI: 10.1016/j.vaccine.2011.08.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 06/16/2011] [Accepted: 08/06/2011] [Indexed: 01/06/2023]
Abstract
The effectiveness of recombinant Adenovirus serotype 5 (Ad5) vectors to induce immune responses against targeted antigens has been limited by the presence of pre-existing or Ad5 vaccine induced anti-vector immunity. The Ad5 [E1-, E2b-] platform, a recombinant Ad5 with additional deletions, has been previously reported by us to induce immune responses in the presence of Ad5 immunity. In an Ad5 immune non-human primate (NHP) model, an Ad5 [E1-, E2b-] construct expressing HIV-1 Gag induced immune responses in the presence of pre-existing Ad5 immunity. In the present study we expand on these prior observations by comparing the cell mediated immune (CMI) responses induced by Ad5 [E1-, E2b-]-SIV-gag/nef in Ad5 naïve and Ad5 immune NHP. Additionally, NHP were immunized with an Ad5 [E1-, E2b-]-HIV-pol construct following two homologous administrations of Ad5 [E1-, E2b-]-SIV-gag/nef to determine if an immune response could be induced against a third antigen in the presence of vaccine induced Ad5 immunity. Positive CMI responses, as assessed by interferon-gamma (IFN-γ) secreting lymphocytes, were induced against all three antigens. These CMI responses increased over a course of multiple immunizations and the response profiles observed in Ad5 naïve and Ad5 immune NHP were similar. No influence of the major histocompatibility complex on CMI responses was observed. These data indicate that the new Ad5 [E1-, E2b-] platform based vaccine could be used for homologous vaccination regimes to induce robust CMI responses in the presence of Ad5 vector immunity.
Collapse
|
49
|
Jones FR, Gabitzsch ES, Xu Y, Balint JP, Borisevich V, Smith J, Smith J, Peng BH, Walker A, Salazar M, Paessler S. Prevention of influenza virus shedding and protection from lethal H1N1 challenge using a consensus 2009 H1N1 HA and NA adenovirus vector vaccine. Vaccine 2011; 29:7020-6. [PMID: 21821082 DOI: 10.1016/j.vaccine.2011.07.073] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 07/07/2011] [Accepted: 07/10/2011] [Indexed: 10/17/2022]
Abstract
Vaccines against emerging pathogens such as the 2009 H1N1 pandemic virus can benefit from current technologies such as rapid genomic sequencing to construct the most biologically relevant vaccine. A novel platform (Ad5 [E1-, E2b-]) has been utilized to induce immune responses to various antigenic targets. We employed this vector platform to express hemagglutinin (HA) and neuraminidase (NA) genes from 2009 H1N1 pandemic viruses. Inserts were consensuses sequences designed from viral isolate sequences and the vaccine was rapidly constructed and produced. Vaccination induced H1N1 immune responses in mice, which afforded protection from lethal virus challenge. In ferrets, vaccination protected from disease development and significantly reduced viral titers in nasal washes. H1N1 cell mediated immunity as well as antibody induction correlated with the prevention of disease symptoms and reduction of virus replication. The Ad5 [E1-, E2b-] should be evaluated for the rapid development of effective vaccines against infectious diseases.
Collapse
|
50
|
Gabitzsch ES, Xu Y, Balcaitis S, Balint JP, Jones FR. An Ad5[E1-, E2b-]-HER2/neu vector induces immune responses and inhibits HER2/neu expressing tumor progression in Ad5 immune mice. Cancer Gene Ther 2011; 18:326-35. [PMID: 21233857 PMCID: PMC3079015 DOI: 10.1038/cgt.2010.82] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Immunotherapy is a promising approach for the treatment of cancers. Modified adenovirus 5 (Ad5) vectors have been used as a platform to deliver genes encoding tumor associated antigens (TAA). A major obstacle to Ad5 vector immunotherapy has been the induction of vector immunity following administration or the presence of pre-existing Ad5 immunity, which results in vector mitigation. It has been reported by us that the Ad5[E1-, E2b-] platform with unique deletions in the E1, E2b and E3 regions can induce potent cell mediated immunity (CMI) against delivered transgene products in the presence of pre-existing Ad5 immunity. Here we report the use of an Ad5[E1-, E2b-] vector platform expressing the TAA HER2/neu as a breast cancer immunotherapeutic agent. Ad5[E1-, E2b-]-HER2/neu induced potent CMI against HER2/neu in Ad5 naïve and Ad5 immune mice. Humoral responses were also induced and antibodies could lyse HER2/neu expressing tumor cells in the presence of complement in vitro. Ad5[E1-, E2b-]-HER2/neu prevented establishment of HER2/neu-expressing tumors and significantly inhibited progression of established tumors in Ad5 naïve and Ad5 immune murine models. These data demonstrate that in vivo delivery of Ad5[E1-, E2b-]-HER2/neu can induce anti-TAA immunity and inhibit progression of HER2/neu expressing cancers.
Collapse
|