1
|
Park S, Cha HN, Shin MG, Park S, Kim Y, Kim MS, Shin KH, Thoudam T, Lee EJ, Wolfe RR, Dan J, Koh JH, Kim IY, Choi I, Lee IK, Sung HK, Park SY. Inhibitory Regulation of FOXO1 in PPARδ Expression Drives Mitochondrial Dysfunction and Insulin Resistance. Diabetes 2024; 73:1084-1098. [PMID: 38656552 DOI: 10.2337/db23-0432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 04/09/2024] [Indexed: 04/26/2024]
Abstract
Forkhead box O1 (FOXO1) regulates muscle growth, but the metabolic role of FOXO1 in skeletal muscle and its mechanisms remain unclear. To explore the metabolic role of FOXO1 in skeletal muscle, we generated skeletal muscle-specific Foxo1 inducible knockout (mFOXO1 iKO) mice and fed them a high-fat diet to induce obesity. We measured insulin sensitivity, fatty acid oxidation, mitochondrial function, and exercise capacity in obese mFOXO1 iKO mice and assessed the correlation between FOXO1 and mitochondria-related protein in the skeletal muscle of patients with diabetes. Obese mFOXO1 iKO mice exhibited improved mitochondrial respiratory capacity, which was followed by attenuated insulin resistance, enhanced fatty acid oxidation, and improved skeletal muscle exercise capacity. Transcriptional inhibition of FOXO1 in peroxisome proliferator-activated receptor δ (PPARδ) expression was confirmed in skeletal muscle, and deletion of PPARδ abolished the beneficial effects of FOXO1 deficiency. FOXO1 protein levels were higher in the skeletal muscle of patients with diabetes and negatively correlated with PPARδ and electron transport chain protein levels. These findings highlight FOXO1 as a new repressor in PPARδ gene expression in skeletal muscle and suggest that FOXO1 links insulin resistance and mitochondrial dysfunction in skeletal muscle via PPARδ. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Soyoung Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Hye-Na Cha
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Min-Gyeong Shin
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Sanghee Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, Republic of Korea
| | - Min-Seob Kim
- Department of Fundamental Environment Research, Environmental Measurement and Analysis Center, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Kyung-Hoon Shin
- Department of Marine Sciences and Convergent Technology, Hanyang University, Ansan, Republic of Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Robert R Wolfe
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Jinmyoung Dan
- Department of Orthopedic Surgery, College of Medicine, CHA University, Gumi, Republic of Korea
| | - Jin-Ho Koh
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Il-Young Kim
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hoon-Ki Sung
- The Hospital for Sick Children Research Institute & Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| |
Collapse
|
2
|
G Popova P, Chen SP, Liao S, Sadarangani M, Blakney AK. Clinical perspective on topical vaccination strategies. Adv Drug Deliv Rev 2024; 208:115292. [PMID: 38522725 DOI: 10.1016/j.addr.2024.115292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/01/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Vaccination is one of the most successful measures in modern medicine to combat diseases, especially infectious diseases, and saves millions of lives every year. Vaccine design and development remains critical and involves many aspects, including the choice of platform, antigen, adjuvant, and route of administration. Topical vaccination, defined herein as the introduction of a vaccine to any of the three layers of the human skin, has attracted interest in recent years as an alternative vaccination approach to the conventional intramuscular administration because of its potential to be needle-free and induce a superior immune response against pathogens. In this review, we describe recent progress in developing topical vaccines, highlight progress in the development of delivery technologies for topical vaccines, discuss potential factors that might impact the topical vaccine efficacy, and provide an overview of the current clinical landscape of topical vaccines.
Collapse
Affiliation(s)
- Petya G Popova
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada
| | - Sunny P Chen
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada
| | - Suiyang Liao
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada; Life Science Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, 950 West 28th Ave, Vancouver, British Columbia V5Z 4H4, Canada; Department of Pediatrics, University of British Columbia, 4480 Oak St, Vancouver, BC V6H 0B3, Canada
| | - Anna K Blakney
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, British Columbia V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, British Columbia V6T 1Z4, Canada.
| |
Collapse
|
3
|
Qi H, Sun Z, Gao T, Yao Y, Wang Y, Li W, Wang X, Wang X, Liu D, Jiang JD. Genetic fusion of CCL11 to antigens enhances antigenicity in nucleic acid vaccines and eradicates tumor mass through optimizing T-cell response. Mol Cancer 2024; 23:46. [PMID: 38459592 PMCID: PMC10921619 DOI: 10.1186/s12943-024-01958-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
Nucleic acid vaccines have shown promising potency and efficacy for cancer treatment with robust and specific T-cell responses. Improving the immunogenicity of delivered antigens helps to extend therapeutic efficacy and reduce dose-dependent toxicity. Here, we systematically evaluated chemokine-fused HPV16 E6/E7 antigen to improve the cellular and humoral immune responses induced by nucleotide vaccines in vivo. We found that fusion with different chemokines shifted the nature of the immune response against the antigens. Although a number of chemokines were able to amplify specific CD8 + T-cell or humoral response alone or simultaneously. CCL11 was identified as the most potent chemokine in improving immunogenicity, promoting specific CD8 + T-cell stemness and generating tumor rejection. Fusing CCL11 with E6/E7 antigen as a therapeutic DNA vaccine significantly improved treatment effectiveness and caused eradication of established large tumors in 92% tumor-bearing mice (n = 25). Fusion antigens with CCL11 expanded the TCR diversity of specific T cells and induced the infiltration of activated specific T cells, neutrophils, macrophages and dendritic cells (DCs) into the tumor, which created a comprehensive immune microenvironment lethal to tumor. Combination of the DNA vaccine with anti-CTLA4 treatment further enhanced the therapeutic effect. In addition, CCL11 could also be used for mRNA vaccine design. To summarize, CCL11 might be a potent T cell enhancer against cancer.
Collapse
Affiliation(s)
- Hailong Qi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China.
- Newish Biological R&D Center, Wuxi, China.
| | - Zhongjie Sun
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
- Newish Biological R&D Center, Wuxi, China
| | - Tianle Gao
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | - Yu Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
- Newish Biological R&D Center, Wuxi, China
| | - Weiwei Li
- Newish Biological R&D Center, Wuxi, China
| | | | | | - Defang Liu
- Newish Biological R&D Center, Wuxi, China
| | - Jian-Dong Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China.
| |
Collapse
|
4
|
Lu B, Lim JM, Yu B, Song S, Neeli P, Sobhani N, K P, Bonam SR, Kurapati R, Zheng J, Chai D. The next-generation DNA vaccine platforms and delivery systems: advances, challenges and prospects. Front Immunol 2024; 15:1332939. [PMID: 38361919 PMCID: PMC10867258 DOI: 10.3389/fimmu.2024.1332939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024] Open
Abstract
Vaccines have proven effective in the treatment and prevention of numerous diseases. However, traditional attenuated and inactivated vaccines suffer from certain drawbacks such as complex preparation, limited efficacy, potential risks and others. These limitations restrict their widespread use, especially in the face of an increasingly diverse range of diseases. With the ongoing advancements in genetic engineering vaccines, DNA vaccines have emerged as a highly promising approach in the treatment of both genetic diseases and acquired diseases. While several DNA vaccines have demonstrated substantial success in animal models of diseases, certain challenges need to be addressed before application in human subjects. The primary obstacle lies in the absence of an optimal delivery system, which significantly hampers the immunogenicity of DNA vaccines. We conduct a comprehensive analysis of the current status and limitations of DNA vaccines by focusing on both viral and non-viral DNA delivery systems, as they play crucial roles in the exploration of novel DNA vaccines. We provide an evaluation of their strengths and weaknesses based on our critical assessment. Additionally, the review summarizes the most recent advancements and breakthroughs in pre-clinical and clinical studies, highlighting the need for further clinical trials in this rapidly evolving field.
Collapse
Affiliation(s)
- Bowen Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Ming Lim
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Boyue Yu
- Department of Environmental Science, Policy, and Management, University of California at Berkeley, Berkeley, CA, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Pavithra K
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
5
|
Kisakov DN, Belyakov IM, Kisakova LA, Yakovlev VA, Tigeeva EV, Karpenko LI. The use of electroporation to deliver DNA-based vaccines. Expert Rev Vaccines 2024; 23:102-123. [PMID: 38063059 DOI: 10.1080/14760584.2023.2292772] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Nucleic acids represent a promising platform for creating vaccines. One disadvantage of this approach is its relatively low immunogenicity. Electroporation (EP) is an effective way to increase the DNA vaccines immunogenicity. However, due to the different configurations of devices used for EP, EP protocols optimization is required not only to enhance immunogenicity, but also to ensure greater safety and tolerability of the EP procedure. AREA COVERED An data analysis for recent years on the DNA vaccines delivery against viral and parasitic infections using EP was carried out. The study of various EP physical characteristics, such as frequency, pulse duration, pulse interval, should be considered along with the immunogenic construct design and the site of delivery of the vaccine, through the study of the immunogenic and protective characteristics of the latter. EXPERT OPINION Future research should focus on regulating the humoral and cellular response required for protection against infectious agents by modifying the EP protocol. Significant efforts will be directed to establishing the possibility of redirecting the immune response toward the Th1 or Th2 response by changing the EP physical parameters. It will allow for an individual selective approach during EP, depending on the pathogen type of an infectious disease.
Collapse
Affiliation(s)
- Denis N Kisakov
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| | - Igor M Belyakov
- Department of medico-biological disciplines, Moscow University for Industry and Finance "Synergy", Moscow, Russia
| | - Lubov A Kisakova
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| | - Vladimir A Yakovlev
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| | - Elena V Tigeeva
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| | - Larisa I Karpenko
- Department of bioengineering, State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Novosibirsk region, Russia
| |
Collapse
|
6
|
Kranjc M, Polajžer T, Novickij V, Miklavčič D. Determination of the Impact of High-Intensity Pulsed Electromagnetic Fields on the Release of Damage-Associated Molecular Pattern Molecules. Int J Mol Sci 2023; 24:14607. [PMID: 37834054 PMCID: PMC10572873 DOI: 10.3390/ijms241914607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
High-Intensity Pulsed Electromagnetic Fields (HI-PEMF) treatment is an emerging noninvasive and contactless alternative to conventional electroporation, since the electric field inside the tissue is induced remotely by an externally applied pulsed magnetic field. Recently, HI-PEMF has been successfully used in the transfer of plasmid DNA and siRNA in vivo, with no or minimal infiltration of immune cells. In addition to gene electrotransfer, treatment with HI-PEMF has also shown potential for electrochemotherapy, where activation of the immune response contributes to the treatment outcome. The immune response can be triggered by immunogenic cell death that is characterized by the release of damage-associated molecular patterns (DAMPs) from damaged or/and dying cells. In this study, the release of the best-known DAMP molecules, i.e., adenosine triphosphate (ATP), calreticulin and high mobility group box 1 protein (HMBG1), after HI-PEMF treatment was investigated in vitro on three different cell lines of different tissue origin and compared with conventional electroporation treatment parameters. We have shown that HI-PEMF by itself does not cause the release of HMGB1 or calreticulin, whereas the release of ATP was detected immediately after HI-PEMF treatment. Our results indicate that HI-PEMF treatment causes no to minimal release of DAMP molecules, which results in minimal/limited activation of the immune response.
Collapse
Affiliation(s)
- Matej Kranjc
- Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, 1000 Ljubljana, Slovenia; (M.K.); (T.P.)
| | - Tamara Polajžer
- Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, 1000 Ljubljana, Slovenia; (M.K.); (T.P.)
| | - Vitalij Novickij
- Institute of High Magnetic Fields, Faculty of Electronics, Vilnius Gediminas Technical University, Plytinės g. 27, 10105 Vilnius, Lithuania;
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Santariskiu g. 5, 08410 Vilnius, Lithuania
| | - Damijan Miklavčič
- Faculty of Electrical Engineering, University of Ljubljana, Trzaska cesta 25, 1000 Ljubljana, Slovenia; (M.K.); (T.P.)
| |
Collapse
|
7
|
Polajžer T, Miklavčič D. Immunogenic Cell Death in Electroporation-Based Therapies Depends on Pulse Waveform Characteristics. Vaccines (Basel) 2023; 11:1036. [PMID: 37376425 DOI: 10.3390/vaccines11061036] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Traditionally, electroporation-based therapies such as electrochemotherapy (ECT), gene electrotransfer (GET) and irreversible electroporation (IRE) are performed with different but typical pulse durations-100 microseconds and 1-50 milliseconds. However, recent in vitro studies have shown that ECT, GET and IRE can be achieved with virtually any pulse duration (millisecond, microsecond, nanosecond) and pulse type (monopolar, bipolar-HFIRE), although with different efficiency. In electroporation-based therapies, immune response activation can affect treatment outcome, and the possibility of controlling and predicting immune response could improve the treatment. In this study, we investigated if different pulse durations and pulse types cause different or similar activations of the immune system by assessing DAMP release (ATP, HMGB1, calreticulin). Results show that DAMP release can be different when different pulse durations and pulse types are used. Nanosecond pulses seems to be the most immunogenic, as they can induce the release of all three main DAMP molecules-ATP, HMGB1 and calreticulin. The least immunogenic seem to be millisecond pulses, as only ATP release was detected and even that assumingly occurs due to increased permeability of the cell membrane. Overall, it seems that DAMP release and immune response in electroporation-based therapies can be controlled though pulse duration.
Collapse
Affiliation(s)
- Tamara Polajžer
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
| | - Damijan Miklavčič
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška 25, 1000 Ljubljana, Slovenia
| |
Collapse
|
8
|
Improved DNA Vaccine Delivery with Needle-Free Injection Systems. Vaccines (Basel) 2023; 11:vaccines11020280. [PMID: 36851159 PMCID: PMC9964240 DOI: 10.3390/vaccines11020280] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
DNA vaccines have inherent advantages compared to other vaccine types, including safety, rapid design and construction, ease and speed to manufacture, and thermostability. However, a major drawback of candidate DNA vaccines delivered by needle and syringe is the poor immunogenicity associated with inefficient cellular uptake of the DNA. This uptake is essential because the target vaccine antigen is produced within cells and then presented to the immune system. Multiple techniques have been employed to boost the immunogenicity and protective efficacy of DNA vaccines, including physical delivery methods, molecular and traditional adjuvants, and genetic sequence enhancements. Needle-free injection systems (NFIS) are an attractive alternative due to the induction of potent immunogenicity, enhanced protective efficacy, and elimination of needles. These advantages led to a milestone achievement in the field with the approval for Restricted Use in Emergency Situation of a DNA vaccine against COVID-19, delivered exclusively with NFIS. In this review, we discuss physical delivery methods for DNA vaccines with an emphasis on commercially available NFIS and their resulting safety, immunogenic effectiveness, and protective efficacy. As is discussed, prophylactic DNA vaccines delivered by NFIS tend to induce non-inferior immunogenicity to electroporation and enhanced responses compared to needle and syringe.
Collapse
|
9
|
Advances of Electroporation-Related Therapies and the Synergy with Immunotherapy in Cancer Treatment. Vaccines (Basel) 2022; 10:vaccines10111942. [PMID: 36423037 PMCID: PMC9692484 DOI: 10.3390/vaccines10111942] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Electroporation is the process of instantaneously increasing the permeability of a cell membrane under a pulsed electric field. Depending on the parameters of the electric pulses and the target cell electrophysiological characteristics, electroporation can be either reversible or irreversible. Reversible electroporation facilitates the delivery of functional genetic materials or drugs to target cells, inducing cell death by apoptosis, mitotic catastrophe, or pseudoapoptosis; irreversible electroporation is an ablative technology which directly ablates a large amount of tissue without causing harmful thermal effects; electrotherapy using an electric field can induce cell apoptosis without any aggressive invasion. Reversible and irreversible electroporation can also activate systemic antitumor immune response and enhance the efficacy of immunotherapy. In this review, we discuss recent progress related to electroporation, and summarize its latest applications. Further, we discuss the synergistic effects of electroporation-related therapies and immunotherapy. We also propose perspectives for further investigating electroporation and immunotherapy in cancer treatment.
Collapse
|
10
|
Translating a Thin-Film Rehydration Method to Microfluidics for the Preparation of a SARS-CoV-2 DNA Vaccine: When Manufacturing Method Matters. Pharmaceutics 2022; 14:pharmaceutics14071427. [PMID: 35890321 PMCID: PMC9316859 DOI: 10.3390/pharmaceutics14071427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 01/21/2023] Open
Abstract
Previous investigations conducted on a liposomal formulation for a SARS-CoV-2 DNA vaccine manufactured using the thin-film layer rehydration method showed promising immunogenicity results in mice. The adaptation of the liposomal formulation to a scalable and reproducible method of manufacture is necessary to continue the investigation of this vaccine candidate. Microfluidics manufacture shows high potential in method translation. The physicochemical characterization of the blank liposomes produced by thin-film layer rehydration or microfluidics were shown to be comparable. However, a difference in lipid nanostructure in the bilayer resulted in a significant difference in the hydration of the thin-film liposomes, ultimately altering their complexation behavior. A study on the complexation of liposomes with the DNA vaccine at various N/P ratios showed different sizes and Zeta-potential values between the two formulations. This difference in the complexation behavior resulted in distinct immunogenicity profiles in mice. The thin-film layer rehydration-manufactured liposomes induced a significantly higher response compared to the microfluidics-manufactured samples. The nanostructural analysis of the two samples revealed the critical importance of understanding the differences between the two formulations that resulted in the different immunogenicity in mice.
Collapse
|
11
|
Trotovšek B, Djokić M, Čemažar M, Serša G. New era of electrochemotherapy in treatment of liver tumors in conjunction with immunotherapies. World J Gastroenterol 2021; 27:8216-8226. [PMID: 35068866 PMCID: PMC8717013 DOI: 10.3748/wjg.v27.i48.8216] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 10/28/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
Electrochemotherapy is a local ablative therapy that increases the cytotoxicity of either bleomycin or cisplatin by applying electric pulses (electroporation) to tumors. It has already been widely used throughout Europe for the treatment of various types of human and veterinary cutaneous tumors, with an objective response rate ranging from 70%-90%, depending on the tumor histotype. Recently, electrochemotherapy was introduced for the treatment of primary liver tumors, such as hepatocellular carcinoma (HCC). The complete response rate was 85% per treated lesion, with a durable response. Therefore, electrochemotherapy could become a treatment of choice for HCC, especially after achieving a transition from an open surgery approach to a percutaneous approach that uses dedicated electrodes. Electrochemotherapy elicits a local immune response and can be considered an in situ vaccination. HCC, among others, is a potentially immunogenic tumor; thus, electrochemotherapy could boost adjuvant immunotherapy to achieve a better and longer-lasting antitumor response. Therefore, therapeutic strategies that combine electrochemotherapy with immune checkpoint inhibitors or adjuvant treatment with cytokines are indicated for HCC. Immunogene therapy using electroporation as a delivery system for plasmid DNA coding for interleukin-12 is a highly promising approach. This electroporation approach has shown efficacy in preclinical settings and veterinary oncology and is awaiting translation for the treatment of liver tumors, i.e., HCC.
Collapse
Affiliation(s)
- Blaž Trotovšek
- Department of Abdominal Surgery, University Medical Centre Ljubljana, Ljubljana 1000, Slovenia
- Medical Faculty Ljubljana, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Mihajlo Djokić
- Department of Abdominal Surgery, University Medical Centre Ljubljana, Ljubljana 1000, Slovenia
- Medical Faculty Ljubljana, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Maja Čemažar
- Department of Experimental Oncology, Institute of Oncology, Ljubljana 1000, Slovenia
- Faculty of Health Sciences, University of Primorska, Izola 6310, Slovenia
| | - Gregor Serša
- Department of Experimental Oncology, Institute of Oncology, Ljubljana 1000, Slovenia
- Faculty of Health Sciences, University of Ljubljana, Ljubljana 1000, Slovenia
| |
Collapse
|
12
|
da Luz JCDS, Antunes F, Clavijo-Salomon MA, Signori E, Tessarollo NG, Strauss BE. Clinical Applications and Immunological Aspects of Electroporation-Based Therapies. Vaccines (Basel) 2021; 9:727. [PMID: 34358144 PMCID: PMC8310106 DOI: 10.3390/vaccines9070727] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022] Open
Abstract
Reversible electropermeabilization (RE) is an ultrastructural phenomenon that transiently increases the permeability of the cell membrane upon application of electrical pulses. The technique was described in 1972 by Neumann and Rosenheck and is currently used in a variety of applications, from medicine to food processing. In oncology, RE is applied for the intracellular transport of chemotherapeutic drugs as well as the delivery of genetic material in gene therapies and vaccinations. This review summarizes the physical changes of the membrane, the particularities of bleomycin, and the immunological aspects involved in electrochemotherapy and gene electrotransfer, two important EP-based cancer therapies in human and veterinary oncology.
Collapse
Affiliation(s)
- Jean Carlos dos Santos da Luz
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, São Paulo 01246-000, Brazil; (J.C.d.S.d.L.); (F.A.); (N.G.T.)
| | - Fernanda Antunes
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, São Paulo 01246-000, Brazil; (J.C.d.S.d.L.); (F.A.); (N.G.T.)
| | | | - Emanuela Signori
- Institute of Translational Pharmacology, CNR, 00133 Rome, Italy;
| | - Nayara Gusmão Tessarollo
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, São Paulo 01246-000, Brazil; (J.C.d.S.d.L.); (F.A.); (N.G.T.)
| | - Bryan E. Strauss
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, São Paulo 01246-000, Brazil; (J.C.d.S.d.L.); (F.A.); (N.G.T.)
| |
Collapse
|
13
|
Batista Napotnik T, Polajžer T, Miklavčič D. Cell death due to electroporation - A review. Bioelectrochemistry 2021; 141:107871. [PMID: 34147013 DOI: 10.1016/j.bioelechem.2021.107871] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/12/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022]
Abstract
Exposure of cells to high voltage electric pulses increases transiently membrane permeability through membrane electroporation. Electroporation can be reversible and is used in gene transfer and enhanced drug delivery but can also lead to cell death. Electroporation resulting in cell death (termed as irreversible electroporation) has been successfully used as a new non-thermal ablation method of soft tissue such as tumours or arrhythmogenic heart tissue. Even though the mechanisms of cell death can influence the outcome of electroporation-based treatments due to use of different electric pulse parameters and conditions, these are not elucidated yet. We review the mechanisms of cell death after electroporation reported in literature, cell injuries that may lead to cell death after electroporation and membrane repair mechanisms involved. The knowledge of membrane repair and cell death mechanisms after cell exposure to electric pulses, targets of electric field in cells need to be identified to optimize existing and develop of new electroporation-based techniques used in medicine, biotechnology, and food technology.
Collapse
Affiliation(s)
- Tina Batista Napotnik
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Tamara Polajžer
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | - Damijan Miklavčič
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia.
| |
Collapse
|
14
|
Komel T, Bosnjak M, Kranjc Brezar S, De Robertis M, Mastrodonato M, Scillitani G, Pesole G, Signori E, Sersa G, Cemazar M. Gene electrotransfer of IL-2 and IL-12 plasmids effectively eradicated murine B16.F10 melanoma. Bioelectrochemistry 2021; 141:107843. [PMID: 34139572 DOI: 10.1016/j.bioelechem.2021.107843] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 05/05/2021] [Accepted: 05/13/2021] [Indexed: 12/18/2022]
Abstract
Gene therapy has become an important approach for treating cancer, and electroporation represents a technology for introducing therapeutic genes into a cell. An example of cancer gene therapy relying on gene electrotransfer is the use of immunomodulatory cytokines, such as interleukin 2 (IL-2) and 12 (IL-12), which directly stimulate immune cells at the tumour site. The aim of our study was to determine the effects of gene electrotransfer with two plasmids encoding IL-2 and IL-12 in vitro and in vivo. Two different pulse protocols, known as EP1 (600 V/cm, 5 ms, 1 Hz, 8 pulses) and EP2 (1300 V/cm, 100 µs, 1 Hz, 8 pulses), were assessed in vitro for application in subsequent in vivo experiments. In the in vivo experiment, gene electrotransfer of pIL-2 and pIL-12 using the EP1 protocol was performed in B16.F10 murine melanoma. Combined treatment of tumours using pIL2 and pIL12 induced significant tumour growth delay and 71% complete tumour regression. Furthermore, in tumours coexpressing IL-2 and IL-12, increased accumulation of dendritic cells and M1 macrophages was obtained along with the activation of proinflammatory signals, resulting in CD4 + and CD8 + T-lymphocyte recruitment and immune memory development in the mice. In conclusion, we demonstrated high antitumour efficacy of combined IL-2 and IL-12 gene electrotransfer protocols in low-immunogenicity murine B16.F10 melanoma.
Collapse
Affiliation(s)
- T Komel
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - M Bosnjak
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia
| | - S Kranjc Brezar
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Medicine, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - M De Robertis
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - M Mastrodonato
- Department of Biology, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - G Scillitani
- Department of Biology, University of Bari, Via Orabona 4, 70126 Bari, Italy
| | - G Pesole
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126 Bari, Italy; National Research Council-Institute of Biomembrane, Bioenergetics, and Molecular Biotechnology (CNR-IBIOM), Via Amendola 122 O, 70126, Bari, Italy
| | - E Signori
- National Research Council-Institute of Translational Pharmacology (CNR-IFT), Via Fosso del Cavaliere 100, Rome, Italy
| | - G Sersa
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Health Sciences, Zdravstvena pot 5, SI - 1000 Ljubljana, Slovenia
| | - M Cemazar
- Institute of Oncology Ljubljana, Department of Experimental Oncology, Zaloska 2, SI-1000 Ljubljana, Slovenia; University of Primorska, Faculty of Health Sciences, Polje 42, SI - 6310 Izola, Slovenia.
| |
Collapse
|
15
|
Long-Term Antitumor CD8 + T Cell Immunity Induced by Endogenously Engineered Extracellular Vesicles. Cancers (Basel) 2021; 13:cancers13092263. [PMID: 34066801 PMCID: PMC8125873 DOI: 10.3390/cancers13092263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The induction of an effective immune response against tumor cells is of a great benefit in the battle against cancers. We recently characterized a novel, safe, and cost-effective strategy to induce an efficient CD8+ T cell immune response against potentially whatever antigen. This technique is based on in vivo engineering of exosomes/extracellular vesicles (EVs), i.e., nanovesicles constitutively released by all healthy cells. Immunogenic EVs are generated by intramuscular injection of a DNA vector expressing an EV-anchoring protein fused with the antigen of interest. In this paper, we applied our vaccine platform to counteract the growth of tumors expressing antigens of Human Papilloma Virus (HPV). We demonstrated that this method is instrumental in curing mice already developing HPV-related tumors. In addition, cured mice were shown to resist a second tumor cell implantation. These results could be of relevance for a possible translation into the clinic of our technology. Abstract We developed an innovative method to induce antigen-specific CD8+ T cytotoxic lymphocyte (CTL) immunity based on in vivo engineering of extracellular vesicles (EVs). This approach employs a DNA vector expressing a mutated HIV-1 Nef protein (Nefmut) deprived of the anti-cellular effects typical of the wild-type isoform, meanwhile showing an unusual efficiency of incorporation into EVs. This function persists even when foreign antigens are fused to its C-terminus. In this way, Nefmut traffics large amounts of antigens fused to it into EVs spontaneously released by the recipient cells. We previously provided evidence that mice injected with a DNA vector expressing the Nefmut/HPV16-E7 fusion protein developed an E7-specific CTL immune response as detected 2 weeks after the second immunization. Here, we extended and optimized the anti-HPV16 CD8+ T cell immune response induced by the endogenously engineered EVs, and evaluated the therapeutic antitumor efficacy over time. We found that the co-injection of DNA vectors expressing Nefmut fused with E6 and E7 generated a stronger anti-HPV16 immune response compared to that observed in mice injected with the single vectors. When HPV16-E6 and -E7 co-expressing tumor cells were implanted before immunization, all mice survived at day 44, whereas no mice injected with either void or Nefmut-expressing vectors survived until day 32 after tumor implantation. A substantial part of immunized mice (7 out of 12) cleared the tumor. When the cured mice were re-challenged with a second tumor cell implantation, none of them developed tumors. Both E6- and E7-specific CD8+ T immunities were still detectable at the end of the observation time. We concluded that the immunity elicited by engineered EVs, besides counteracting and curing already developed tumors, was strong enough to guarantee the resistance to additional tumor attacks. These results can be of relevance for the therapy of both metastatic and relapsing tumors.
Collapse
|
16
|
Paston SJ, Brentville VA, Symonds P, Durrant LG. Cancer Vaccines, Adjuvants, and Delivery Systems. Front Immunol 2021; 12:627932. [PMID: 33859638 PMCID: PMC8042385 DOI: 10.3389/fimmu.2021.627932] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
Vaccination was first pioneered in the 18th century by Edward Jenner and eventually led to the development of the smallpox vaccine and subsequently the eradication of smallpox. The impact of vaccination to prevent infectious diseases has been outstanding with many infections being prevented and a significant decrease in mortality worldwide. Cancer vaccines aim to clear active disease instead of aiming to prevent disease, the only exception being the recently approved vaccine that prevents cancers caused by the Human Papillomavirus. The development of therapeutic cancer vaccines has been disappointing with many early cancer vaccines that showed promise in preclinical models often failing to translate into efficacy in the clinic. In this review we provide an overview of the current vaccine platforms, adjuvants and delivery systems that are currently being investigated or have been approved. With the advent of immune checkpoint inhibitors, we also review the potential of these to be used with cancer vaccines to improve efficacy and help to overcome the immune suppressive tumor microenvironment.
Collapse
Affiliation(s)
| | | | - Peter Symonds
- Biodiscovery Institute, Scancell Limited, Nottingham, United Kingdom
| | - Lindy G. Durrant
- Biodiscovery Institute, University of Nottingham, Faculty of Medicine and Health Sciences, Nottingham, United Kingdom
| |
Collapse
|
17
|
Han KH, Jang MS, Han HY, Im WJ, Jung KJ, Park KS, Choi D, Jeong HG, Kim SK, Moon KS. Preclinical safety assessment of a therapeutic human papillomavirus DNA vaccine combined with intravaginal interleukin-7 fused with hybrid Fc in female rats. Toxicol Appl Pharmacol 2021; 413:115406. [PMID: 33434572 DOI: 10.1016/j.taap.2021.115406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 01/03/2021] [Accepted: 01/07/2021] [Indexed: 11/19/2022]
Abstract
This study was conducted to establish the toxicological profile of combination treatment with therapeutic HPV DNA vaccines (GX-188E) and the long-acting form of recombinant human interleukin-7 fused with hybrid Fc (IL-7hyFc). GX-188E was administered intramuscularly by electroporation with or without IL-7hyFc intravaginally once per 2 weeks for 8 weeks (five times) in female Sprague-Dawley rats. Because up-regulation of immune responses and migration of antigen-specific T cells in cervicoviginal tissue were predicted as therapeutic effects, we distinguished adverse effects from therapeutic effects based on the severity of the systemic immune response, reversibility of lymphoid tissue changes, target tissue damage, and off-target immune responses. We observed that the number of neutrophils was increased, and the number of lymphocytes was decreased in the blood. Further, myofiber degeneration, necrosis, fibroplasia, and cell infiltration were observed at the GX-188E administration site. These changes were fully or partially recovered over a 4-week period. Analysis of lymphocytes in spleen revealed that CD4+ T cells and total T cells decreased in rats treated with GX-188E in combination with a high dose of IL-7hyFc (1.25 mg/animal). However, these changes were not considered adverse because they were transient and may have been related to electroporation-mediated DNA delivery or the local migration of lymphocytes induced by IL-7. Therefore, the potential toxicity of the combination of GX-188E and IL-7hyFc treatment was comparable to that of GX-188E treatment alone, and the no observed adverse effect level for GX-188E with IL-7hyFc was considered as 320 μg/animal for GX-188E and 1.25 mg/animal for IL-7hyFc.
Collapse
Affiliation(s)
- Kang-Hyun Han
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Min Seong Jang
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea; Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Hyoung-Yun Han
- Department of Predictive Toxicology, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Wan-Jung Im
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Kyung Jin Jung
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Ki Seok Park
- Research Institute, Genexine Inc., Korea Bio Park, Seongnam, Gyeonggi-do 13488, Republic of Korea
| | - Donghoon Choi
- Research Institute, NeoImmunetech, Inc., Rm501-1 Uspace2 B, Seongnam, Gyeonggi-do 13494, Republic of Korea
| | - Hye Gwang Jeong
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| | - Kyoung-Sik Moon
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34114, Republic of Korea.
| |
Collapse
|
18
|
Maglietti F, Tellado M, De Robertis M, Michinski S, Fernández J, Signori E, Marshall G. Electroporation as the Immunotherapy Strategy for Cancer in Veterinary Medicine: State of the Art in Latin America. Vaccines (Basel) 2020; 8:E537. [PMID: 32957424 PMCID: PMC7564659 DOI: 10.3390/vaccines8030537] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Electroporation is a technology that increases cell membrane permeability by the application of electric pulses. Electrochemotherapy (ECT), the best-known application of electroporation, is a very effective local treatment for tumors of any histology in human and veterinary medicine. It induces a local yet robust immune response that is responsible for its high effectiveness. Gene electrotransfer (GET), used in research to produce a systemic immune response against cancer, is another electroporation-based treatment that is very appealing for its effectiveness, low cost, and simplicity. In this review, we present the immune effect of electroporation-based treatments and analyze the results of the vast majority of the published papers related to immune response enhancement by gene electrotransfer in companion animals with spontaneous tumors. In addition, we present a brief history of the initial steps and the state of the art of the electroporation-based treatments in Latin America. They have the potential to become an essential form of immunotherapy in the region. This review gives insight into the subject and helps to choose promising research lines for future work; it also helps to select the adequate treatment parameters for performing a successful application of this technology.
Collapse
Affiliation(s)
- Felipe Maglietti
- Instituto Universitario del Hospital Italiano de Buenos Aires, CONICET, Buenos Aires 1199, Argentina
| | - Matías Tellado
- VetOncologia, Veterinary Oncology Clinic, Buenos Aires 1408, Argentina; (M.T.); (J.F.)
| | - Mariangela De Robertis
- CNR-Institute of Biomembrane, Bioenergetics, and Molecular Biotechnology, 70126 Bari, Italy;
- Department of Bioscience, Biotechnology, and Biopharmaceutics, University of Bari, 70126 Bari, Italy
| | - Sebastián Michinski
- Instituto de Física del Plasma, DF, FCEyN, UBA-CONICET, Buenos Aires 1428, Argentina; (S.M.); (G.M.)
| | - Juan Fernández
- VetOncologia, Veterinary Oncology Clinic, Buenos Aires 1408, Argentina; (M.T.); (J.F.)
| | - Emanuela Signori
- Laboratory of Molecular Pathology and Experimental Oncology, Institute of Translational Pharmacology, CNR, 00133 Rome, Italy;
| | - Guillermo Marshall
- Instituto de Física del Plasma, DF, FCEyN, UBA-CONICET, Buenos Aires 1428, Argentina; (S.M.); (G.M.)
| |
Collapse
|
19
|
Analysis of damage-associated molecular pattern molecules due to electroporation of cells in vitro. Radiol Oncol 2020; 54:317-328. [PMID: 32726295 PMCID: PMC7409611 DOI: 10.2478/raon-2020-0047] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/07/2020] [Indexed: 01/10/2023] Open
Abstract
Background Tumor cells can die via immunogenic cell death pathway, in which damage-associated molecular pattern molecules (DAMPs) are released from the cells. These molecules activate cells involved in the immune response. Both innate and adaptive immune response can be activated, causing a destruction of the remaining infected cells. Activation of immune response is also an important component of tumor treatment with electrochemotherapy (ECT) and irreversible electroporation (IRE). We thus explored, if and when specific DAMPs are released as a consequence of electroporation in vitro. Materials and methods In this in vitro study, 100 μs long electric pulses were applied to a suspension of Chinese hamster ovary cells. The release of DAMPs - specifically: adenosine triphosphate (ATP), calreticulin, nucleic acids and uric acid was investigated at different time points after exposing the cells to electric pulses of different amplitudes. The release of DAMPs was statistically correlated with cell permeabilization and cell survival, e.g. reversible and irreversible electroporation. Results In general, the release of DAMPs increases with increasing pulse amplitude. Concentration of DAMPs depend on the time interval between exposure of the cells to pulses and the analysis. Concentrations of most DAMPs correlate strongly with cell death. However, we detected no uric acid in the investigated samples. Conclusions Release of DAMPs can serve as a marker for prediction of cell death. Since the stability of certain DAMPs is time dependent, this should be considered when designing protocols for detecting DAMPs after electric pulse treatment.
Collapse
|
20
|
Sokołowska E, Błachnio-Zabielska AU. A Critical Review of Electroporation as A Plasmid Delivery System in Mouse Skeletal Muscle. Int J Mol Sci 2019; 20:ijms20112776. [PMID: 31174257 PMCID: PMC6600476 DOI: 10.3390/ijms20112776] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
The gene delivery to skeletal muscles is a promising strategy for the treatment of both muscular disorders (by silencing or overexpression of specific gene) and systemic secretion of therapeutic proteins. The use of a physical method like electroporation with plate or needle electrodes facilitates long-lasting gene silencing in situ. It has been reported that electroporation enhances the expression of the naked DNA gene in the skeletal muscle up to 100 times and decreases the changeability of the intramuscular expression. Coelectransfer of reporter genes such as green fluorescent protein (GFP), luciferase or beta-galactosidase allows the observation of correctly performed silencing in the muscles. Appropriate selection of plasmid injection volume and concentration, as well as electrotransfer parameters, such as the voltage, the length and the number of electrical pulses do not cause long-term damage to myocytes. In this review, we summarized the electroporation methodology as well as the procedure of electrotransfer to the gastrocnemius, tibialis, soleus and foot muscles and compare their advantages and disadvantages.
Collapse
Affiliation(s)
- Emilia Sokołowska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 15-222 Bialystok, Poland.
| | | |
Collapse
|
21
|
Zhang R, Billingsley MM, Mitchell MJ. Biomaterials for vaccine-based cancer immunotherapy. J Control Release 2018; 292:256-276. [PMID: 30312721 PMCID: PMC6355332 DOI: 10.1016/j.jconrel.2018.10.008] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/06/2018] [Accepted: 10/08/2018] [Indexed: 12/28/2022]
Abstract
The development of therapeutic cancer vaccines as a means to generate immune reactivity against tumors has been explored since the early discovery of tumor-specific antigens by Georg Klein in the 1960s. However, challenges including weak immunogenicity, systemic toxicity, and off-target effects of cancer vaccines remain as barriers to their broad clinical translation. Advances in the design and implementation of biomaterials are now enabling enhanced efficacy and reduced toxicity of cancer vaccines by controlling the presentation and release of vaccine components to immune cells and their microenvironment. Here, we discuss the rational design and clinical status of several classes of cancer vaccines (including DNA, mRNA, peptide/protein, and cell-based vaccines) along with novel biomaterial-based delivery technologies that improve their safety and efficacy. Further, strategies for designing new platforms for personalized cancer vaccines are also considered.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Margaret M Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
22
|
De Robertis M, Pasquet L, Loiacono L, Bellard E, Messina L, Vaccaro S, Di Pasquale R, Fazio VM, Rols MP, Teissie J, Golzio M, Signori E. In Vivo Evaluation of a New Recombinant Hyaluronidase to Improve Gene Electro-Transfer Protocols for DNA-Based Drug Delivery against Cancer. Cancers (Basel) 2018; 10:cancers10110405. [PMID: 30373297 PMCID: PMC6265783 DOI: 10.3390/cancers10110405] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 10/08/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022] Open
Abstract
Cancer vaccines based on plasmid DNA represent a good therapeutic perspective, despite their low potency. Animal-derived hyaluronidases (Hyals) are employed in oncological clinical practice. Hyal has been also demonstrated to be a good enhancer of intramuscular Gene Electro-Transfer (GET) efficiency in anti-cancer preclinical protocols, with increased transfected cells and higher expression of the encoded genes. Nevertheless, the use of animal-derived Hyals results limited respect to their potentialities, since such preparations could be affected by low purity, variable potency and uncertain safety. To improve the delivery of intramuscular GET-based protocols in mouse, we investigated a new recombinant Hyal, the rHyal-sk, to assess in vivo safety and activity of this treatment at cellular and biochemical levels. We evaluated the cellular events and the inflammation chemical mediators involved at different time points after rHyal-sk administration plus GET. Our results demonstrated the in vivo safety and efficacy of rHyal-sk when injected once intramuscularly in association with GET, with no toxicity, good plasmid in-take ability, useful inflammatory response activation, and low immunogenicity. Following these findings, we would recommend the use of the new rHyal-sk for the delivery of DNA-based vaccines and immunotherapy, as well as into clinical practice, for tumor disease treatments.
Collapse
Affiliation(s)
- Mariangela De Robertis
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari "A. Moro", via Orabona 4, 70126 Bari, Italy.
- Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, via Alvaro del Portillo 21, 00128 Rome, Italy.
- CNR-Institute of Translational Pharmacology, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| | - Lise Pasquet
- Vaccine Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA.
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, BP64182, 205 Route de Narbonne, 31077 Toulouse, France.
| | - Luisa Loiacono
- Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, via Alvaro del Portillo 21, 00128 Rome, Italy.
- New Drug Modalities, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, UK.
| | - Elisabeth Bellard
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, BP64182, 205 Route de Narbonne, 31077 Toulouse, France.
| | - Luciano Messina
- Fidia Farmaceutici S.p.A., Local Unit Fidia Research Sud, Contrada Pizzuta snc, 96017 Noto, Siracusa, Italy.
| | - Susanna Vaccaro
- Fidia Farmaceutici S.p.A., Local Unit Fidia Research Sud, Contrada Pizzuta snc, 96017 Noto, Siracusa, Italy.
| | - Roberta Di Pasquale
- Fidia Farmaceutici S.p.A., Local Unit Fidia Research Sud, Contrada Pizzuta snc, 96017 Noto, Siracusa, Italy.
| | - Vito Michele Fazio
- Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, via Alvaro del Portillo 21, 00128 Rome, Italy.
- Fondazione IRCCS Casa Sollievo della Sofferenza, Laboratorio di Oncologia, viale dei Cappuccini, 71013 San Giovanni Rotondo (FG), Italy.
| | - Marie-Pierre Rols
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, BP64182, 205 Route de Narbonne, 31077 Toulouse, France.
| | - Justin Teissie
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, BP64182, 205 Route de Narbonne, 31077 Toulouse, France.
| | - Muriel Golzio
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, BP64182, 205 Route de Narbonne, 31077 Toulouse, France.
| | - Emanuela Signori
- Laboratory of Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, via Alvaro del Portillo 21, 00128 Rome, Italy.
- CNR-Institute of Translational Pharmacology, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| |
Collapse
|
23
|
Brentville VA, Atabani S, Cook K, Durrant LG. Novel tumour antigens and the development of optimal vaccine design. Ther Adv Vaccines Immunother 2018; 6:31-47. [PMID: 29998219 DOI: 10.1177/2515135518768769] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 02/23/2018] [Indexed: 12/13/2022] Open
Abstract
The interplay between tumours and the immune system has long been known to involve complex interactions between tumour cells, immune cells and the tumour microenvironment. The progress of checkpoint inhibitors in the clinic in the last decade has highlighted again the role of the immune system in the fight against cancer. Numerous efforts have been undertaken to develop ways of stimulating the cellular immune response to eradicate tumours. These interventions include the identification of appropriate tumour antigens as targets for therapy. In this review, we summarize progress in selection of target tumour antigen. Targeting self antigens has the problem of thymic deletion of high-affinity T-cell responses leaving a diminished repertoire of low-affinity T cells that fail to kill tumour cells. Thymic regulation appears to be less stringent for differentiation of cancer-testis antigens, as many tumour rejection antigens fall into this category. More recently, targeting neo-epitopes or post-translational modifications such as a phosphorylation or stress-induced citrullination has shown great promise in preclinical studies. Of particular interest is that the responses can be mediated by both CD4 and CD8 T cells. Previous vaccines have targeted CD8 T-cell responses but more recently, the central role of CD4 T cells in orchestrating inflammation within tumours and also differentiating into potent killer cells has been recognized. The design of vaccines to induce such immune responses is discussed herein. Liposomally encoded ribonucleic acid (RNA), targeted deoxyribonucleic acid (DNA) or long peptides linked to toll-like receptor (TLR) adjuvants are the most promising new vaccine approaches. These exciting new approaches suggest that the 'Holy Grail' of a simple nontoxic cancer vaccine may be on the horizon. A major hurdle in tumour therapy is also to overcome the suppressive tumour environment. We address current progress in combination therapies and suggest that these are likely to show the most promise for the future.
Collapse
Affiliation(s)
| | - Suha Atabani
- Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Katherine Cook
- Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Lindy G Durrant
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital, Hucknall Road, Nottingham, NG5 1PB, UK
| |
Collapse
|
24
|
Patel PM, Ottensmeier CH, Mulatero C, Lorigan P, Plummer R, Pandha H, Elsheikh S, Hadjimichael E, Villasanti N, Adams SE, Cunnell M, Metheringham RL, Brentville VA, Machado L, Daniels I, Gijon M, Hannaman D, Durrant LG. Targeting gp100 and TRP-2 with a DNA vaccine: Incorporating T cell epitopes with a human IgG1 antibody induces potent T cell responses that are associated with favourable clinical outcome in a phase I/II trial. Oncoimmunology 2018; 7:e1433516. [PMID: 29872563 PMCID: PMC5980353 DOI: 10.1080/2162402x.2018.1433516] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 02/08/2023] Open
Abstract
A DNA vaccine, SCIB1, incorporating two CD8 and two CD4 epitopes from TRP-2/gp100 was evaluated in patients with metastatic melanoma. Each patient received SCIB1 via intramuscular injection with electroporation. The trial was designed to find the safest dose of SCIB1 which induced immune/clinical responses in patients with or without tumour. Fifteen patients with tumor received SCIB1 doses of 0.4-8 mg whilst 20 fully-resected patients received 2-8 mg doses. Twelve patients elected to continue immunization every 3 months for up to 39 months. SCIB1 induced dose-dependent T cell responses in 88% of patients with no serious adverse effects or dose limiting toxicities. The intensity of the T cell responses was significantly higher in patients receiving 4 mg doses without tumor when compared to those with tumor (p < 0.01). In contrast, patients with tumor showed a significantly higher response to the 8 mg dose than the 4 mg dose (p < 0.03) but there was no significant difference in the patients without tumor. One of 15 patients with measurable disease showed an objective tumor response and 7/15 showed stable disease. 5/20 fully-resected patients have experienced disease recurrence but all remained alive at the cut-off date with a median observation time of 37 months. A positive clinical outcome was associated with MHC-I and MHC-II expression on tumors prior to therapy (p = 0.027). We conclude that SCIB1 is well tolerated and stimulates potent T cell responses in melanoma patients. It deserves further evaluation as a single agent adjuvant therapy or in combination with checkpoint inhibitors in advanced disease.
Collapse
Affiliation(s)
- Poulam M Patel
- Academic Department of Clinical Oncology, Division of Cancer & Stem Cells, University of Nottingham, Nottingham, UK
| | - Christian H Ottensmeier
- Southampton Experimental Cancer Medicine Centre and Southampton University Hospitals, Faculty of Medicine, Southampton, UK
| | | | - Paul Lorigan
- Institute of Cancer Sciences, University of Manchester, The Christie NHS Foundation Trust, Manchester, UK
| | - Ruth Plummer
- Northern Institute for Cancer Research, Medical School, University of Newcastle-upon-Tyne and Wear, UK
| | - Hardev Pandha
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Somaia Elsheikh
- University of Nottingham, School of Medicine Queen's Medical Centre, Nottingham, UK
| | | | - Naty Villasanti
- University of Nottingham, School of Medicine Queen's Medical Centre, Nottingham, UK
| | - Sally E Adams
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Michelle Cunnell
- Academic Department of Clinical Oncology, Division of Cancer & Stem Cells, University of Nottingham, Nottingham, UK
| | - Rachael L Metheringham
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Victoria A Brentville
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Lee Machado
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Ian Daniels
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | - Mohamed Gijon
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| | | | - Lindy G Durrant
- Academic Department of Clinical Oncology, Division of Cancer & Stem Cells, University of Nottingham, Nottingham, UK.,Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, Nottingham, UK
| |
Collapse
|
25
|
Adipose tissue: a new target for electroporation-enhanced DNA vaccines. Gene Ther 2017; 24:757-767. [PMID: 29106403 PMCID: PMC5746593 DOI: 10.1038/gt.2017.96] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/29/2017] [Accepted: 10/11/2017] [Indexed: 12/20/2022]
Abstract
DNA vaccines delivered using electroporation (EP) have had clinical success, but these EP methods generally utilize invasive needle electrodes. Here, we demonstrate the delivery and immunogenicity of a DNA vaccine into subcutaneous adipose tissue cells using noninvasive EP. Using finite element analysis, we predicted that plate electrodes, when oriented properly, could effectively concentrate the electric field within adipose tissue. In practice, these electrodes generated widespread gene expression persisting for at least 60 days in vivo within interscapular subcutaneous fat pads of guinea pigs. We then applied this adipose-EP protocol to deliver a DNA vaccine coding for an influenza antigen into guinea pigs. The resulting host immune responses elicited were of a similar magnitude to those achieved by skin delivery with EP. The onset of the humoral immune response was more rapid when the DNA dose was spread over multiple injection sites, and increasing the voltage of the EP device increased the magnitude of the immune response. This study supports further development of EP protocols delivering gene-based therapies to subcutaneous fat.
Collapse
|
26
|
Gene Electrotransfer of Plasmid-Encoding IL-12 Recruits the M1 Macrophages and Antigen-Presenting Cells Inducing the Eradication of Aggressive B16F10 Murine Melanoma. Mediators Inflamm 2017; 2017:5285890. [PMID: 28596641 PMCID: PMC5449735 DOI: 10.1155/2017/5285890] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/03/2017] [Accepted: 03/12/2017] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy is currently one of the leading approaches in cancer treatment. Gene electrotransfer of plasmids encoding interleukin 12 (IL-12) into the cells leads to the production of IL-12, which drives immune cell polarization to an antitumoral response. One of the cell types that shows great promise in targeting tumor cells under the influence of IL-12 cytokine milieu is that of macrophages. Therefore, the aim of this study was to evaluate gene electrotransfer of antibiotic resistance-free plasmid DNA-encoding murine IL-12 (mIL-12) in mice bearing aggressive B16F10 murine melanoma. IL-12 electrotransfer resulted in the complete long-term eradication of the tumors. Serum mIL-12 and murine interferon γ (mIFNγ) were increased after IL-12 gene electrotransfer. Further on, hematoxylin and eosin (HE) staining showed increased infiltration of immune cells that lasted from day 4 until day 14. Immunohistochemistry (IHC) staining of F4/80, MHCII, and CD11c showed higher positive staining in the IL-12 gene electrotransfer group than in the control groups. Immune cell infiltration into the tumors and the high density of MHCII- and CD11c-positive cells suggest an antitumor polarization of macrophages and the presence of antigen-presenting cells that contributes to the important antitumor effectiveness of IL-12.
Collapse
|
27
|
Bagley KC, Schwartz JA, Andersen H, Eldridge JH, Xu R, Ota-Setlik A, Geltz JJ, Halford WP, Fouts TR. An Interleukin 12 Adjuvanted Herpes Simplex Virus 2 DNA Vaccine Is More Protective Than a Glycoprotein D Subunit Vaccine in a High-Dose Murine Challenge Model. Viral Immunol 2017; 30:178-195. [PMID: 28085634 DOI: 10.1089/vim.2016.0136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Vaccination is a proven intervention against human viral diseases; however, success against Herpes Simplex Virus 2 (HSV-2) remains elusive. Most HSV-2 vaccines tested in humans to date contained just one or two immunogens, such as the virion attachment receptor glycoprotein D (gD) and/or the envelope fusion protein, glycoprotein B (gB). At least three factors may have contributed to the failures of subunit-based HSV-2 vaccines. First, immune responses directed against one or two viral antigens may lack sufficient antigenic breadth for efficacy. Second, the antibody responses elicited by these vaccines may have lacked necessary Fc-mediated effector functions. Third, these subunit vaccines may not have generated necessary protective cellular immune responses. We hypothesized that a polyvalent combination of HSV-2 antigens expressed from a DNA vaccine with an adjuvant that polarizes immune responses toward a T helper 1 (Th1) phenotype would compose a more effective vaccine. We demonstrate that delivery of DNA expressing full-length HSV-2 glycoprotein immunogens by electroporation with the adjuvant interleukin 12 (IL-12) generates substantially greater protection against a high-dose HSV-2 vaginal challenge than a recombinant gD subunit vaccine adjuvanted with alum and monophosphoryl lipid A (MPL). Our results further show that DNA vaccines targeting optimal combinations of surface glycoproteins provide better protection than gD alone and provide similar survival benefits and disease symptom reductions compared with a potent live attenuated HSV-2 0ΔNLS vaccine, but that mice vaccinated with HSV-2 0ΔNLS clear the virus much faster. Together, our data indicate that adjuvanted multivalent DNA vaccines hold promise for an effective HSV-2 vaccine, but that further improvements may be required.
Collapse
Affiliation(s)
| | | | | | | | - Rong Xu
- 3 Profectus Biosciences , Tarrytown, New York
| | | | - Joshua J Geltz
- 4 Department of Microbiology and Immunology, Southern Illinois University School of Medicine , Springfield, Illinois
| | - William P Halford
- 4 Department of Microbiology and Immunology, Southern Illinois University School of Medicine , Springfield, Illinois
| | | |
Collapse
|
28
|
Kostrzak A, Caval V, Escande M, Pliquet E, Thalmensi J, Bestetti T, Julithe M, Fiette L, Huet T, Wain-Hobson S, Langlade-Demoyen P. APOBEC3A intratumoral DNA electroporation in mice. Gene Ther 2016; 24:74-83. [PMID: 27858943 DOI: 10.1038/gt.2016.77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 10/26/2016] [Accepted: 11/11/2016] [Indexed: 12/21/2022]
Abstract
Human APOBEC3A (A3A) cytidine deaminase shows pro-apoptotic properties resulting from hypermutation of genomic DNA, induction of double-stranded DNA breaks (DSBs) and G1 cell cycle arrest. Given this, we evaluated the antitumor efficacy of A3A by intratumoral electroporation of an A3A expression plasmid. DNA was repeatedly electroporated into B16OVA, B16Luc tumors of C57BL/6J mice as well as the aggressive fibrosarcoma Sarc2 tumor of HLA-A*0201/DRB1*0101 transgenic mice using noninvasive plate electrodes. Intratumoral electroporation of A3A plasmid DNA resulted in regression of ~50% of small B16OVA melanoma tumors that did not rebound in the following 2 months without treatment. Larger or more aggressive tumors escaped regression when so treated. As APOBEC3A was much less efficient in provoking hypermutation and DSBs in B16OVA cells compared with human or quail cells, it is likely that APOBEC3A would be more efficient in a human setting than in a mouse model.
Collapse
Affiliation(s)
- A Kostrzak
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - V Caval
- Molecular Retrovirology Unit, Institut Pasteur, Paris, France
| | - M Escande
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - E Pliquet
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - J Thalmensi
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - T Bestetti
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - M Julithe
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - L Fiette
- Human Histopathology and Animal Models, Infection & Epidemiology Department, Institut Pasteur, Paris, France.,Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
| | - T Huet
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France
| | - S Wain-Hobson
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France.,Molecular Retrovirology Unit, Institut Pasteur, Paris, France
| | - P Langlade-Demoyen
- Invectys, Pepinière Paris Biotech Santé Cochin, Paris, France.,Molecular Retrovirology Unit, Institut Pasteur, Paris, France
| |
Collapse
|
29
|
Jorritsma SHT, Gowans EJ, Grubor-Bauk B, Wijesundara DK. Delivery methods to increase cellular uptake and immunogenicity of DNA vaccines. Vaccine 2016; 34:5488-5494. [PMID: 27742218 DOI: 10.1016/j.vaccine.2016.09.062] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/20/2016] [Accepted: 09/29/2016] [Indexed: 12/22/2022]
Abstract
DNA vaccines are ideal candidates for global vaccination purposes because they are inexpensive and easy to manufacture on a large scale such that even people living in low-income countries can benefit from vaccination. However, the potential of DNA vaccines has not been realized owing mainly to the poor cellular uptake of DNA in vivo resulting in the poor immunogenicity of DNA vaccines. In this review, we discuss the benefits and shortcomings of several promising and innovative non-biological methods of DNA delivery that can be used to increase cellular delivery and efficacy of DNA vaccines.
Collapse
Affiliation(s)
- S H T Jorritsma
- Virology Research Group, Discipline of Surgery, The Basil Hetzel Institute, The University of Adelaide, Australia
| | - E J Gowans
- Virology Research Group, Discipline of Surgery, The Basil Hetzel Institute, The University of Adelaide, Australia
| | - B Grubor-Bauk
- Virology Research Group, Discipline of Surgery, The Basil Hetzel Institute, The University of Adelaide, Australia
| | - D K Wijesundara
- Virology Research Group, Discipline of Surgery, The Basil Hetzel Institute, The University of Adelaide, Australia.
| |
Collapse
|
30
|
Lambricht L, Vanvarenberg K, De Beuckelaer A, Van Hoecke L, Grooten J, Ucakar B, Lipnik P, Sanders NN, Lienenklaus S, Préat V, Vandermeulen G. Coadministration of a Plasmid Encoding HIV-1 Gag Enhances the Efficacy of Cancer DNA Vaccines. Mol Ther 2016; 24:1686-96. [PMID: 27434590 DOI: 10.1038/mt.2016.122] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 06/09/2016] [Indexed: 02/07/2023] Open
Abstract
DNA vaccination holds great promise for the prevention and treatment of cancer and infectious diseases. However, the clinical ability of DNA vaccines is still controversial due to the limited immune response initially observed in humans. We hypothesized that electroporation of a plasmid encoding the HIV-1 Gag viral capsid protein would enhance cancer DNA vaccine potency. DNA electroporation used to deliver plasmids in vivo, induced type I interferons, thereby supporting the activation of innate immunity. The coadministration of ovalbumin (OVA) and HIV-1 Gag encoding plasmids modulated the adaptive immune response. This strategy favored antigen-specific Th1 immunity, delayed B16F10-OVA tumor growth and improved mouse survival in both prophylactic and therapeutic vaccination approaches. Similarly, a prophylactic DNA immunization against the melanoma-associated antigen gp100 was enhanced by the codelivery of the HIV-1 Gag plasmid. The adjuvant effect was not driven by the formation of HIV-1 Gag virus-like particles. This work highlights the ability of both electroporation and the HIV-1 Gag plasmid to stimulate innate immunity for enhancing cancer DNA vaccine immunogenicity and demonstrates interesting tracks for the design of new translational genetic adjuvants to overcome the current limitations of DNA vaccines in humans.
Collapse
Affiliation(s)
- Laure Lambricht
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Kevin Vanvarenberg
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Ans De Beuckelaer
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,VIB Medical Biotechnology Center, Ghent University, Ghent, Belgium
| | - Johan Grooten
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bernard Ucakar
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Pascale Lipnik
- Bio and Soft Matter, Université catholique de Louvain, Louvain-La-Neuve, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany.,Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Hannover, Germany
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Gaëlle Vandermeulen
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
31
|
Xue W, Metheringham RL, Brentville VA, Gunn B, Symonds P, Yagita H, Ramage JM, Durrant LG. SCIB2, an antibody DNA vaccine encoding NY-ESO-1 epitopes, induces potent antitumor immunity which is further enhanced by checkpoint blockade. Oncoimmunology 2016; 5:e1169353. [PMID: 27471648 PMCID: PMC4938367 DOI: 10.1080/2162402x.2016.1169353] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/07/2016] [Accepted: 03/17/2016] [Indexed: 01/11/2023] Open
Abstract
Checkpoint blockade has demonstrated promising antitumor responses in approximately 10-40% of patients. However, the majority of patients do not make a productive immune response to their tumors and do not respond to checkpoint blockade. These patients may benefit from an effective vaccine that stimulates high-avidity T cell responses in combination with checkpoint blockade. We have previously shown that incorporating TRP-2 and gp100 epitopes into the CDR regions of a human IgG1 DNA (ImmunoBody®: IB) results in significant tumor regression both in animal models and patients. This vaccination strategy is superior to others as it targets antigen to antigen-presenting cells and stimulates high-avidity T cell responses. To broaden the application of this vaccination strategy, 16 NY-ESO-1 epitopes, covering over 80% of HLA phenotypes, were incorporated into the IB (SCIB2). They produced higher frequency and avidity T cell responses than peptide vaccination. These T cells were of sufficient avidity to kill NY-ESO-1-expressing tumor cells, and in vivo controlled the growth of established B16-NY-ESO-1 tumors, resulting in long-term survival (35%). When SCIB2 was given in combination with Treg depletion, CTLA-4 blockade or PD-1 blockade, long-term survival from established tumors was significantly enhanced to 56, 67 and 100%, respectively. Translating these responses into the clinic by using a combination of SCIB2 vaccination and checkpoint blockade can only further improve clinical responses.
Collapse
Affiliation(s)
- Wei Xue
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus , Nottingham, UK
| | - Rachael L Metheringham
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus , Nottingham, UK
| | - Victoria A Brentville
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus , Nottingham, UK
| | - Barbara Gunn
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus , Nottingham, UK
| | - Peter Symonds
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus , Nottingham, UK
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine , Tokyo, Japan
| | - Judith M Ramage
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus , Nottingham, UK
| | - Lindy G Durrant
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK; Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottingham, UK
| |
Collapse
|
32
|
Abstract
Plasmids are currently an indispensable molecular tool in life science research and a central asset for the modern biotechnology industry, supporting its mission to produce pharmaceutical proteins, antibodies, vaccines, industrial enzymes, and molecular diagnostics, to name a few key products. Furthermore, plasmids have gradually stepped up in the past 20 years as useful biopharmaceuticals in the context of gene therapy and DNA vaccination interventions. This review provides a concise coverage of the scientific progress that has been made since the emergence of what are called today plasmid biopharmaceuticals. The most relevant topics are discussed to provide researchers with an updated overview of the field. A brief outline of the initial breakthroughs and innovations is followed by a discussion of the motivation behind the medical uses of plasmids in the context of therapeutic and prophylactic interventions. The molecular characteristics and rationale underlying the design of plasmid vectors as gene transfer agents are described and a description of the most important methods used to deliver plasmid biopharmaceuticals in vivo (gene gun, electroporation, cationic lipids and polymers, and micro- and nanoparticles) is provided. The major safety issues (integration and autoimmunity) surrounding the use of plasmid biopharmaceuticals is discussed next. Aspects related to the large-scale manufacturing are also covered, and reference is made to the plasmid products that have received marketing authorization as of today.
Collapse
|
33
|
Lambricht L, Lopes A, Kos S, Sersa G, Préat V, Vandermeulen G. Clinical potential of electroporation for gene therapy and DNA vaccine delivery. Expert Opin Drug Deliv 2015; 13:295-310. [PMID: 26578324 DOI: 10.1517/17425247.2016.1121990] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Electroporation allows efficient delivery of DNA into cells and tissues, thereby improving the expression of therapeutic or immunogenic proteins that are encoded by plasmid DNA. This simple and versatile method holds a great potential and could address unmet medical needs such as the prevention or treatment of many cancers or infectious diseases. AREAS COVERED This review explores the electroporation mechanism and the parameters affecting its efficacy. An analysis of past and current clinical trials focused on DNA electroporation is presented. The pathologies addressed, the protocol used, the treatment outcome and the tolerability are highlighted. In addition, several of the possible optimization strategies for improving patient compliance and therapeutic efficacy are discussed such as plasmid design, use of genetic adjuvants for DNA vaccines, choice of appropriate delivery site and electrodes as well as pulse parameters. EXPERT OPINION The growing number of clinical trials and the results already available underline the strong potential of DNA electroporation which combines both safety and efficiency. Nevertheless, it remains critical to further increase fundamental knowledge to refine future strategies, to develop concerted and common DNA electroporation protocols and to continue exploring new electroporation-based therapeutic options.
Collapse
Affiliation(s)
- Laure Lambricht
- a Université catholique de Louvain, Louvain Drug Research Institute , Advanced Drug Delivery and Biomaterials , Brussels , Belgium
| | - Alessandra Lopes
- a Université catholique de Louvain, Louvain Drug Research Institute , Advanced Drug Delivery and Biomaterials , Brussels , Belgium
| | - Spela Kos
- b Institute of Oncology Ljubljana , Department of Experimental Oncology , Ljubljana , Slovenia
| | - Gregor Sersa
- b Institute of Oncology Ljubljana , Department of Experimental Oncology , Ljubljana , Slovenia
| | - Véronique Préat
- a Université catholique de Louvain, Louvain Drug Research Institute , Advanced Drug Delivery and Biomaterials , Brussels , Belgium
| | - Gaëlle Vandermeulen
- a Université catholique de Louvain, Louvain Drug Research Institute , Advanced Drug Delivery and Biomaterials , Brussels , Belgium
| |
Collapse
|
34
|
Sersa G, Teissie J, Cemazar M, Signori E, Kamensek U, Marshall G, Miklavcic D. Electrochemotherapy of tumors as in situ vaccination boosted by immunogene electrotransfer. Cancer Immunol Immunother 2015; 64:1315-27. [PMID: 26067277 PMCID: PMC4554735 DOI: 10.1007/s00262-015-1724-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/26/2015] [Indexed: 12/21/2022]
Abstract
Electroporation is a platform technology for drug and gene delivery. When applied to cell in vitro or tissues in vivo, it leads to an increase in membrane permeability for molecules which otherwise cannot enter the cell (e.g., siRNA, plasmid DNA, and some chemotherapeutic drugs). The therapeutic effectiveness of delivered chemotherapeutics or nucleic acids depends greatly on their successful and efficient delivery to the target tissue. Therefore, the understanding of different principles of drug and gene delivery is necessary and needs to be taken into account according to the specificity of their delivery to tumors and/or normal tissues. Based on the current knowledge, electrochemotherapy (a combination of drug and electric pulses) is used for tumor treatment and has shown great potential. Its local effectiveness is up to 80 % of local tumor control, however, without noticeable effect on metastases. In an attempt to increase systemic antitumor effectiveness of electrochemotherapy, electrotransfer of genes with immunomodulatory effect (immunogene electrotransfer) could be used as adjuvant treatment. Since electrochemotherapy can induce immunogenic cell death, adjuvant immunogene electrotransfer to peritumoral tissue could lead to locoregional effect as well as the abscopal effect on distant untreated metastases. Therefore, we propose a combination of electrochemotherapy with peritumoral IL-12 electrotransfer, as a proof of principle, using electrochemotherapy boosted with immunogene electrotransfer as in situ vaccination for successful tumor treatment.
Collapse
Affiliation(s)
- Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, 1000, Ljubljana, Slovenia,
| | | | | | | | | | | | | |
Collapse
|
35
|
Glebova K, Reznik ON, Reznik AO, Mehta R, Galkin A, Baranova A, Skoblov M. siRNA technology in kidney transplantation: current status and future potential. BioDrugs 2015; 28:345-61. [PMID: 24573958 DOI: 10.1007/s40259-014-0087-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Kidney transplantation is one of the most common transplantation operations in the world, accounting for up to 50 % of all transplantation surgeries. To curtail the damage to transplanted organs that is caused by ischemia-reperfusion injury and the recipient's immune system, small interfering RNA (siRNA) technology is being explored. Importantly, the kidney as a whole is a preferential site for non-specific systemic delivery of siRNA. To date, most attempts at siRNA-based therapy for transplantation-related conditions have remained at the in vitro stage, with only a few of them being advanced into animal models. Hydrodynamic intravenous injection of naked or carrier-bound siRNAs is currently the most common route for delivery of therapeutic constructs. To our knowledge, no systematic screens for siRNA targets most relevant for kidney transplantation have been attempted so far. A majority of researchers have arrived at one or another target of interest by analyzing current literature that dissects pathological processes taking place in transplanted organs. A majority of the genes that make up the list of 53 siRNA targets that have been tested in transplantation-related models so far belong to either apoptosis- or immune rejection-centered networks. There is an opportunity for therapeutic siRNA combinations that may be delivered within the same delivery vector or injected at the same time and, by targeting more than one pathway, or by hitting the same pathways within two different key points, will augment the effects of each other.
Collapse
Affiliation(s)
- Kristina Glebova
- Research Center for Medical Genetics, Russian Academy of Medical Sciences, Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
36
|
Kos S, Tesic N, Kamensek U, Blagus T, Cemazar M, Kranjc S, Lavrencak J, Sersa G. Improved Specificity of Gene Electrotransfer to Skin Using pDNA Under the Control of Collagen Tissue-Specific Promoter. J Membr Biol 2015; 248:919-28. [PMID: 25840832 DOI: 10.1007/s00232-015-9799-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/26/2015] [Indexed: 11/26/2022]
Abstract
In order to ensure safe, efficient and controlled gene delivery to skin, the improvement of delivery methods together with proper design of DNA is required. Non-viral delivery methods, such as gene electrotransfer, and the design of tissue-specific promoters are promising tools to ensure the safety of gene delivery to the skin. In the scope of our study, we evaluated a novel skin-specific plasmid DNA with collagen (COL) promoter, delivered to skin cells and skin tissue by gene electrotransfer. In vitro, we determined the specificity of the COL promoter in fibroblast cells. The specific expression under the control of COL promoter was obtained for the reporter gene DsRed as well as for therapeutic gene encoding cytokine IL-12. In vivo, the plasmid with COL promoter encoding the reporter gene DsRed was efficiently transfected to mouse skin. It resulted in the notable and controlled manner, however, in lower and shorter expression, compared to that obtained with ubiquitous promoter. The concentration of the IL-12 in the skin after the in vivo transfection of plasmid with COL promoter was in the same range as after the treatment in control conditions (injection of distilled water followed by the application of electric pulses). Furthermore, this gene delivery was local, restricted to the skin, without any evident systemic shedding of IL-12. Such specific targeting of skin cells, observed with tissue-specific COL promoter, would improve the effectiveness and safety of cutaneous gene therapies and DNA vaccines.
Collapse
Affiliation(s)
- Spela Kos
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, 1000, Ljubljana, Slovenia
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol 2015; 35 Suppl:S185-S198. [PMID: 25818339 DOI: 10.1016/j.semcancer.2015.03.004] [Citation(s) in RCA: 1069] [Impact Index Per Article: 106.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 12/27/2022]
Abstract
Cancer immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Although considerable progress has been made in understanding how cancers evade destructive immunity, measures to counteract tumor escape have not kept pace. There are a number of factors that contribute to tumor persistence despite having a normal host immune system. Immune editing is one of the key aspects why tumors evade surveillance causing the tumors to lie dormant in patients for years through "equilibrium" and "senescence" before re-emerging. In addition, tumors exploit several immunological processes such as targeting the regulatory T cell function or their secretions, antigen presentation, modifying the production of immune suppressive mediators, tolerance and immune deviation. Besides these, tumor heterogeneity and metastasis also play a critical role in tumor growth. A number of potential targets like promoting Th1, NK cell, γδ T cell responses, inhibiting Treg functionality, induction of IL-12, use of drugs including phytochemicals have been designed to counter tumor progression with much success. Some natural agents and phytochemicals merit further study. For example, use of certain key polysaccharide components from mushrooms and plants have shown to possess therapeutic impact on tumor-imposed genetic instability, anti-growth signaling, replicative immortality, dysregulated metabolism etc. In this review, we will discuss the advances made toward understanding the basis of cancer immune evasion and summarize the efficacy of various therapeutic measures and targets that have been developed or are being investigated to enhance tumor rejection.
Collapse
|
38
|
Olaiz N, Signori E, Maglietti F, Soba A, Suárez C, Turjanski P, Michinski S, Marshall G. Tissue damage modeling in gene electrotransfer: The role of pH. Bioelectrochemistry 2014; 100:105-11. [DOI: 10.1016/j.bioelechem.2014.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 04/22/2014] [Accepted: 05/02/2014] [Indexed: 02/05/2023]
|
39
|
Mahindhoratep S, Bouda HA, El Shafey N, Scherman D, Kichler A, Pichon C, Midoux P, Mignet N, Bureau MF. NF-kB related transgene expression in mouse tibial cranial muscle after pDNA injection followed or not by electrotransfer. Biochim Biophys Acta Gen Subj 2014; 1840:3257-63. [PMID: 24973564 DOI: 10.1016/j.bbagen.2014.06.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/28/2014] [Accepted: 06/18/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND When activated, NF-κB can promote the nuclear import and transcription of DNA possessing NF-κB consensus sequences. Here, we investigated whether NF-κB is involved in the plasmid electrotransfer process. METHODS Mouse tibial cranial muscles were transfected with plasmids encoding luciferase bearing or not NF-κB consensus sequences. Luciferase transgene expression was evaluated noninvasively by luminescence imaging and the number of pDNA copies in the same muscles by qPCR. RT-PCR of heat shock protein HsP70 mRNA evidenced cell stress. Western blots of phosphorylated IkBα were studied as a marker of NF-κB activation. RESULTS Intra-muscular injection of a plasmid bearing a weak TATA-like promoter results in a very low muscle transfection level. Electrotransfer significantly increased both the number of pDNA copy and the transgene expression of this plasmid per DNA copy. Insertion of NF-κB consensus sequences into pDNA significantly increased the level of gene expression both with and without electrotransfer. Electrotransfer-induced cellular stress was evidenced by increased HsP70 mRNA. Phosphorylated IκBα was slightly increased by simple pDNA injection and a little more by electrotransfer. We also observed a basal level of phosphorylated IκBα and thus of free NF-κB in the absence of any stimulation. GENERAL SIGNIFICANCE pDNA electrotransfer can increase transgene expression independently of NF-κB. The insertion of NF-κB consensus sequences into pDNA bearing a weak TATA-like promoter leads to enhanced transgene expression in muscle with or without gene electrotransfer. Finally, our results suggest that the basal amount of free NF-κB in muscle might be sufficient to enhance the activity of pDNA bearing NF-κB consensus sequences.
Collapse
Affiliation(s)
- S Mahindhoratep
- U1022 INSERM, UMR8258 CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé, Chimie ParisTech, Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - H Ait Bouda
- U1022 INSERM, UMR8258 CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé, Chimie ParisTech, Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - Nelly El Shafey
- U1022 INSERM, UMR8258 CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé, Chimie ParisTech, Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - D Scherman
- U1022 INSERM, UMR8258 CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé, Chimie ParisTech, Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - A Kichler
- V-SAT, UMR7199 CNRS, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch Cedex, France
| | - Ch Pichon
- UPR4301 CNRS, rue Charles Sadron, 45071 Orléans Cedex 02, France
| | - P Midoux
- UPR4301 CNRS, rue Charles Sadron, 45071 Orléans Cedex 02, France
| | - N Mignet
- U1022 INSERM, UMR8258 CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé, Chimie ParisTech, Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - M F Bureau
- U1022 INSERM, UMR8258 CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé, Chimie ParisTech, Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006 Paris, France
| |
Collapse
|
40
|
Short noncoding DNA fragments improve the immune potency of electroporation-mediated HBV DNA vaccination. Gene Ther 2014; 21:703-8. [DOI: 10.1038/gt.2014.44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 03/11/2014] [Accepted: 04/01/2014] [Indexed: 12/11/2022]
|
41
|
Calvet CY, André FM, Mir LM. Dual therapeutic benefit of electroporation-mediated DNA vaccination in vivo: Enhanced gene transfer and adjuvant activity. Oncoimmunology 2014; 3:e28540. [PMID: 25050220 PMCID: PMC4077865 DOI: 10.4161/onci.28540] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 03/14/2014] [Indexed: 01/08/2023] Open
Abstract
DNA vaccination consists of administering an antigen-coding nucleotide sequence. In order to improve the efficacy of DNA vaccines, electroporation is one of the most commonly used methods to enhance DNA uptake. Here, we discuss additional immunological effects of electroporation that are key aspects for inducing immunity in response to DNA vaccines.
Collapse
Affiliation(s)
- Christophe Y Calvet
- Univ Paris-Sud; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France ; CNRS; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France ; Gustave Roussy; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France
| | - Franck M André
- Univ Paris-Sud; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France ; CNRS; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France ; Gustave Roussy; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France
| | - Lluis M Mir
- Univ Paris-Sud; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France ; CNRS; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France ; Gustave Roussy; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses; UMR 8203; Villejuif, France
| |
Collapse
|
42
|
Ragonnaud E, Holst P. The rationale of vectored gene-fusion vaccines against cancer: evolving strategies and latest evidence. THERAPEUTIC ADVANCES IN VACCINES 2014; 1:33-47. [PMID: 24757514 DOI: 10.1177/2051013613480446] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of vaccines that target tumor antigens in cancer has proven difficult. A major reason for this is that T cells specific for tumor self-antigens and neoantigens are eliminated or inactivated through mechanisms of tolerance. Antigen fusion strategies which increase the ability of vaccines to stimulate T cells that have escaped tolerance mechanisms, may have a particular potential as immunotherapies. This review highlights antigen fusion strategies that have been successful in stimulating the induction of T-cell immunity against cancer and counteracting tumor-associated tolerance. In preclinical studies, these strategies have shown to improve the potency of vectored vaccines through fusion of tumor antigen to proteins or protein domains that increase CD4+ T-cell help, CD8+ T-cell responses or both the CD4+ and CD8+ T-cell responses. However, in clinical trials such strategies seem to be less efficient when provided as a DNA vaccine. The first clinical trial using a viral vectored fusion-gene vaccine is expected to be tested as a partner in a heterologous prime-boost regimen directed against cervical cancer.
Collapse
Affiliation(s)
| | - Peter Holst
- ISIM - Center for Medical Parasitology, Copenhagen, Denmark
| |
Collapse
|
43
|
Calvet CY, Famin D, André FM, Mir LM. Electrochemotherapy with bleomycin induces hallmarks of immunogenic cell death in murine colon cancer cells. Oncoimmunology 2014; 3:e28131. [PMID: 25083316 PMCID: PMC4111940 DOI: 10.4161/onci.28131] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/04/2014] [Accepted: 02/06/2014] [Indexed: 01/08/2023] Open
Abstract
Electrochemotherapy (ECT) is a local cancer treatment that has been used over the course of more than 2 decades for the removal of cutaneous and subcutaneous tumors. Several lines of evidence support the premise that the immune system is an important factor underlying anticancer treatment efficacy, potentially including patient responses to ECT. The concept of immunogenic cell death (ICD) arose a few years ago, stating that some cancer treatments generate danger-associated molecular patterns (DAMPs) that trigger an adaptive immune response against tumors. Hence, dying cancer cells behave as a therapeutic vaccine, eliciting a cytotoxic immune response against surviving malignant cells. In our study, we sought to evaluate the ability of ECT to generate cancer cell death encompassing the immunostimulatory characteristics of ICD. To this end, we assayed CT26 murine colon cancer cells in vitro in response to either electric pulses (EPs) application only or in combination with the anticancer drug bleomycin (that is ECT) by quantification of calreticulin (CRT) membrane externalization, as well as the liberation of adenosine triphosphate (ATP) and high mobility group box 1 (HMGB1) protein. We show here that cell permeabilizing yet non-lethal electric pulses induce CRT exposure on the cell surface of EP-only treated cancer cells, as well as ATP release. However, the association of electric pulses along with the chemotherapeutic agent bleomycin was mandatory for HMGB1 release coincident with regimen-induced cell death. These data obtained in vitro were then substantiated by vaccination protocols performed in immunocompetent mice, showing that the injection of dying ECT-treated cells elicits an antitumor immune response that prevents the growth of a subsequent administration of viable cancer cells. We also confirmed previous results showing ECT treatment is much more efficient in immunocompetent animals than in immunodeficient ones, causing complete regressions in the former but not in the latter. This supports a central role for immunity in this beneficial outcome. In conclusion, we show that ECT not only possesses an intrinsic cytotoxic property toward cancer cells but also generates a systemic anticancer immune response via the activation of ICD. Hence, ECT may represent an interesting approach to treat solid tumors while preventing recurrence and metastasis, possibly in combination with immunostimulating agents.
Collapse
Affiliation(s)
- Christophe Y Calvet
- Univ Paris-Sud; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France ; CNRS, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France ; Gustave Roussy; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France
| | - Delphine Famin
- Univ Paris-Sud; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France ; CNRS, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France ; Gustave Roussy; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France
| | - Franck M André
- Univ Paris-Sud; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France ; CNRS, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France ; Gustave Roussy; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France
| | - Lluis M Mir
- Univ Paris-Sud; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France ; CNRS, Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France ; Gustave Roussy; Laboratoire de Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203; Villejuif, France
| |
Collapse
|
44
|
Enhanced immunogenicity of an HIV-1 DNA vaccine delivered with electroporation via combined intramuscular and intradermal routes. J Virol 2014; 88:6959-69. [PMID: 24719412 DOI: 10.1128/jvi.00183-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED It is accepted that an effective prophylactic HIV-1 vaccine is likely to have the greatest impact on viral transmission rates. As previous reports have implicated DNA-priming, protein boost regimens to be efficient activators of humoral responses, we sought to optimize this regimen to further augment vaccine immunogenicity. Here we evaluated single versus concurrent intradermal (i.d.) and intramuscular (i.m.) vaccinations as a DNA-priming strategy for their abilities to elicit humoral and cellular responses against a model HIV-1 vaccine antigen, CN54-gp140. To further augment vaccine-elicited T and B cell responses, we enhanced cellular transfection with electroporation and then boosted the DNA-primed responses with homologous protein delivered subcutaneously (s.c.), intranasally (i.n.), i.m., or transcutaneously (t.c.). In mice, the concurrent priming regimen resulted in significantly elevated gamma interferon T cell responses and high-avidity antigen-specific IgG B cell responses, a hallmark of B cell maturation. Protein boosting of the concurrent DNA strategy further enhanced IgG concentrations but had little impact on T cell reactivity. Interestingly protein boosting by the subcutaneous route increased antibody avidity to a greater extent than protein boosting by either the i.m., i.n., or t.c. route, suggesting that this route may be preferential for driving B cell maturation. Using an alternative and larger animal model, the rabbit, we found the concurrent DNA-priming strategy followed by s.c. protein boosting to again be capable of eliciting high-avidity humoral responses and to also be able to neutralize HIV-1 pseudoviruses from diverse clades (clades A, B, and C). Taken together, we show that concurrent multiple-route DNA vaccinations induce strong cellular immunity, in addition to potent and high-avidity humoral immune responses. IMPORTANCE The route of vaccination has profound effects on prevailing immune responses. Due to the insufficient immunogenicity and protection of current DNA delivery strategies, we evaluated concurrent DNA delivery via simultaneous administration of plasmid DNA by the i.m. and i.d. routes. The rationale behind this study was to provide clear evidence of the utility of concurrent vaccinations for an upcoming human clinical trial. Furthermore, this work will guide future preclinical studies by evaluating the use of model antigens and plasmids for prime-boost strategies. This paper will be of interest not only to virologists and vaccinologists working in the HIV field but also to researchers working in other viral vaccine settings and, critically, to the wider field of vaccine delivery.
Collapse
|
45
|
van Drunen Littel-van den Hurk S, Hannaman D. Electroporation for DNA immunization: clinical application. Expert Rev Vaccines 2014; 9:503-17. [DOI: 10.1586/erv.10.42] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
46
|
Chiarella P, Signori E. Intramuscular DNA vaccination protocols mediated by electric fields. Methods Mol Biol 2014; 1121:315-24. [PMID: 24510835 DOI: 10.1007/978-1-4614-9632-8_28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vaccination is historically one of the most important methods for preventing infectious diseases in humans and animals. New insights in the biology of the immune system allow a more rational design of vaccines, and new vaccination strategies are emerging. DNA vaccines have been proposed as a promising approach for introducing foreign antigens into the host for inducing protective immunity against infectious and cancer diseases. Nevertheless, because of their poor immunogenicity, plasmid DNA vaccination strategies need further implementations. Recent data suggest electrotransfer as a useful tool to improve DNA-based vaccination protocols, being able to stimulate both the humoral and cellular immune responses. In preclinical trials, gene electrotransfer is successfully used in prime-boost combination protocols and its tolerability and safety has been demonstrated also in Phase I clinical trials. In this chapter, we report a short comment supporting electrotransfer as an effective strategy to improve DNA-based vaccination protocols and describe the vaccination procedures by plasmid DNA in combination with electrotransfer and hyaluronidase pretreatment in use in our laboratory.
Collapse
Affiliation(s)
- Pieranna Chiarella
- Laboratory of Molecular Pathology and Experimental Oncology, CNR-IFT, Rome, Italy
| | | |
Collapse
|
47
|
Induction of antigen-positive cell death by the expression of perforin, but not DTa, from a DNA vaccine enhances the immune response. Immunol Cell Biol 2013; 92:359-67. [PMID: 24323081 DOI: 10.1038/icb.2013.93] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/28/2013] [Accepted: 11/09/2013] [Indexed: 02/06/2023]
Abstract
The failure of traditional protein-based vaccines to prevent infection by viruses such as HIV or hepatitis C highlights the need for novel vaccine strategies. DNA vaccines have shown promise in small animal models, and are effective at generating anti-viral T cell-mediated immune responses; however, they have proved to be poorly immunogenic in clinical trials. We propose that the induction of necrosis will enhance the immune response to vaccine antigens encoded by DNA vaccines, as necrotic cells are known to release a range of intracellular factors that lead to dendritic cell (DC) activation and enhanced cross-presentation of antigen. Here we provide evidence that induction of cell death in DNA vaccine-targeted cells provides an adjuvant effect following intradermal vaccination of mice; however, this enhancement of the immune response is dependent on both the mechanism and timing of cell death after antigen expression. We report that a DNA vaccine encoding the cytolytic protein, perforin, resulted in DC activation, enhanced broad and multifunctional CD8 T-cell responses to the HIV-1 antigen GAG and reduced viral load following challenge with a chimeric virus, EcoHIV, compared with the canonical GAG DNA vaccine. This effect was not observed for a DNA vaccine encoding an apoptosis-inducing toxin, DTa, or when the level of perforin expression was increased to induce cell death sooner after vaccination. Thus, inducing lytic cell death following a threshold level of expression of a viral antigen can improve the immunogenicity of DNA vaccines, whereas apoptotic cell death has an inhibitory effect on the immune response.
Collapse
|
48
|
In vivo electroporation of minicircle DNA as a novel method of vaccine delivery to enhance HIV-1-specific immune responses. J Virol 2013; 88:1924-34. [PMID: 24284319 DOI: 10.1128/jvi.02757-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
DNA vaccines offer advantage over conventional vaccines, as they are safer to use, easier to produce, and able to induce humoral as well cellular immune responses. Unfortunately, no DNA vaccines have been licensed for human use for the difficulties in developing an efficient and safe in vivo gene delivery system. In vivo electroporation (EP)-based DNA delivery has attracted great attention for its potency to enhance cellular uptake of DNA vaccines and function as an adjuvant. Minicircle DNA (a new form of DNA containing only a gene expression cassette and lacking a backbone of bacterial plasmid DNA) is a powerful candidate of gene delivery in terms of improving the levels and the duration of transgene expression in vivo. In this study, as a novel vaccine delivery system, we combined in vivo EP and the minicircle DNA carrying a codon-optimized HIV-1 gag gene (minicircle-gag) to evaluate the immunogenicity of this system. We found that minicircle-gag conferred persistent and high levels of gag expression in vitro and in vivo. The use of EP delivery further increased minicircle-based gene expression. Moreover, when delivered by EP, minicircle-gag vaccination elicited a 2- to 3-fold increase in cellular immune response and a 1.5- to 3-fold augmentation of humoral immune responses compared with those elicited by a pVAX1-gag positive control. Increased immunogenicity of EP-assisted minicircle-gag may benefit from increasing local antigen expression, upregulating inflammatory genes, and recruiting immune cells. Collectively, in vivo EP of minicircle DNA functions as a novel vaccine platform that can enhance efficacy and immunogenicity of DNA vaccines.
Collapse
|
49
|
Cu Y, Broderick KE, Banerjee K, Hickman J, Otten G, Barnett S, Kichaev G, Sardesai NY, Ulmer JB, Geall A. Enhanced Delivery and Potency of Self-Amplifying mRNA Vaccines by Electroporation in Situ. Vaccines (Basel) 2013; 1:367-83. [PMID: 26344119 PMCID: PMC4494232 DOI: 10.3390/vaccines1030367] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/12/2013] [Accepted: 08/14/2013] [Indexed: 01/06/2023] Open
Abstract
Nucleic acid-based vaccines such as viral vectors, plasmid DNA (pDNA), and mRNA are being developed as a means to address limitations of both live-attenuated and subunit vaccines. DNA vaccines have been shown to be potent in a wide variety of animal species and several products are now licensed for commercial veterinary but not human use. Electroporation delivery technologies have been shown to improve the generation of T and B cell responses from synthetic DNA vaccines in many animal species and now in humans. However, parallel RNA approaches have lagged due to potential issues of potency and production. Many of the obstacles to mRNA vaccine development have recently been addressed, resulting in a revival in the use of non-amplifying and self-amplifying mRNA for vaccine and gene therapy applications. In this paper, we explore the utility of EP for the in vivo delivery of large, self-amplifying mRNA, as measured by reporter gene expression and immunogenicity of genes encoding HIV envelope protein. These studies demonstrated that EP delivery of self-amplifying mRNA elicited strong and broad immune responses in mice, which were comparable to those induced by EP delivery of pDNA.
Collapse
Affiliation(s)
- Yen Cu
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | | | - Kaustuv Banerjee
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Julie Hickman
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Gillis Otten
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Susan Barnett
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Gleb Kichaev
- Inovio Pharmaceuticals, Blue Bell, PA 19422, USA
| | | | - Jeffrey B Ulmer
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Andrew Geall
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA.
| |
Collapse
|
50
|
Kichaev G, Mendoza JM, Amante D, Smith TRF, McCoy JR, Sardesai NY, Broderick KE. Electroporation mediated DNA vaccination directly to a mucosal surface results in improved immune responses. Hum Vaccin Immunother 2013; 9:2041-8. [PMID: 23954979 DOI: 10.4161/hv.25272] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In vivo electroporation (EP) has been shown to be a highly efficient non-viral method for enhancing DNA vaccine delivery and immunogenicity, when the site of immunization is the skin or muscle of animals and humans. However, the route of entry for many microbial pathogens is via the mucosal surfaces of the human body. We have previously reported on minimally invasive, surface and contactless EP devices for enhanced DNA delivery to dermal tissue. Robust antibody responses were induced following vaccine delivery in several tested animal models using these devices. Here, we investigated extending the modality of the surface device to efficiently deliver DNA vaccines to mucosal tissue. Initially, we demonstrated reporter gene expression in the epithelial layer of buccal mucosa in a guinea pig model. There was minimal tissue damage in guinea pig mucosal tissue resulting from EP. Delivery of a DNA vaccine encoding influenza virus nucleoprotein (NP) of influenza H1N1 elicited robust and sustained systemic IgG antibody responses following EP-enhanced delivery in the mucosa. Upon further analysis, IgA antibody responses were detected in vaginal washes and sustained cellular immune responses were detected in animals immunized at the oral mucosa with the surface EP device. This data confirms that DNA delivery and EP targeting mucosal tissue directly results in both robust and sustainable humoral as well as cellular immune responses without tissue damage. These responses are seen both in the mucosa and systemically in the blood. Direct DNA vaccine delivery enhanced by EP in mucosa may have important clinical applications for delivery of prophylactic and therapeutic DNA vaccines against diseases such as HIV, HPV and pneumonia that enter at mucosal sites and require both cellular and humoral immune responses for protection.
Collapse
Affiliation(s)
| | | | | | | | - Jay R McCoy
- Inovio Pharmaceuticals Inc.;Blue Bell, PA USA
| | | | | |
Collapse
|