1
|
Han M, Nguyen B, Lee JY, Browning E, Zhang J, Mukhopadhyay A, Gujar R, Salazar J, Hermiz R, Svenson L, Rolig AS, Redmond WL, Algazi AP, Daud AI, Canton DA, Twitty CG. Intratumoral electroporation of plasmid encoded IL-12 and membrane-anchored anti-CD3 increases systemic tumor immunity. Mol Cancer Res 2022; 20:983-995. [PMID: 35302641 DOI: 10.1158/1541-7786.mcr-21-0834] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/15/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022]
Abstract
Intratumoral delivery of plasmid IL 12 via electroporation (IT tavo EP) induces localized expression of IL 12 leading to regression of treated and distant tumors with durable responses and minimal toxicity. A key driver in amplifying this local therapy into a systemic response is the magnitude and composition of immune infiltrate in the treated tumor. While intratumoral IL 12 typically increases the density of CD3+ tumor infiltrating lymphocytes (TIL), this infiltrate is composed of a broad range of T cell subsets, including activated tumor specific T cells, less functional bystander T cells, as well as suppressive T regulatory cells. To encourage a more favorable on treatment tumor microenvironment, we explored combining this IL 12 therapy with an intratumoral polyclonal T cell stimulator membrane anchored anti CD3 to productively engage a diverse subset of lymphocytes including the non reactive and suppressive T cells. This study highlighted that combined intratumoral electroporation of IL 12 and membrane anchored anti CD3 plasmids can enhance cytokine production, T cell cytotoxicity, and proliferation while limiting the suppressive capacity within the TME. These collective anti tumor effects not only improve regression of treated tumors but drive systemic immunity with control of non treated contralateral tumors in vivo. Moreover, combination of IL 12 and anti CD3 restored the function of TIL isolated from a melanoma patient actively progressing on PD 1 checkpoint inhibitor therapy. This DNA encodable polyclonal T cell stimulator (membrane anchored anti CD3 plasmid) may represent a key addition to intratumoral IL-12 therapies in the clinic.
Collapse
Affiliation(s)
- Mia Han
- OncoSec Medical Inc., San Diego, United States
| | | | - Jack Y Lee
- OncoSec Medical Inc., San Diego, CA, United States
| | | | - Jun Zhang
- Oncosec Medical Inc., San Diego, CA, United States
| | | | | | - Jon Salazar
- Oncosec Medical Inc., San Diego, CA, United States
| | | | | | - Annah S Rolig
- Providence Cancer Institute, Portland, OR, United States
| | | | - Alain P Algazi
- University of California, San Francisco, San Francisco, CA, United States
| | - Adil I Daud
- University of California, San Francisco, San Francisco, CA, United States
| | - David A Canton
- Oncosec Medical Incorporated, San Diego, CA, United States
| | | |
Collapse
|
2
|
Glassman CR, Mathiharan YK, Jude KM, Su L, Panova O, Lupardus PJ, Spangler JB, Ely LK, Thomas C, Skiniotis G, Garcia KC. Structural basis for IL-12 and IL-23 receptor sharing reveals a gateway for shaping actions on T versus NK cells. Cell 2021; 184:983-999.e24. [PMID: 33606986 PMCID: PMC7899134 DOI: 10.1016/j.cell.2021.01.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 11/23/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
Interleukin-12 (IL-12) and IL-23 are heterodimeric cytokines that are produced by antigen-presenting cells to regulate the activation and differentiation of lymphocytes, and they share IL-12Rβ1 as a receptor signaling subunit. We present a crystal structure of the quaternary IL-23 (IL-23p19/p40)/IL-23R/IL-12Rβ1 complex, together with cryoelectron microscopy (cryo-EM) maps of the complete IL-12 (IL-12p35/p40)/IL-12Rβ2/IL-12Rβ1 and IL-23 receptor (IL-23R) complexes, which reveal "non-canonical" topologies where IL-12Rβ1 directly engages the common p40 subunit. We targeted the shared IL-12Rβ1/p40 interface to design a panel of IL-12 partial agonists that preserved interferon gamma (IFNγ) induction by CD8+ T cells but impaired cytokine production from natural killer (NK) cells in vitro. These cell-biased properties were recapitulated in vivo, where IL-12 partial agonists elicited anti-tumor immunity to MC-38 murine adenocarcinoma absent the NK-cell-mediated toxicity seen with wild-type IL-12. Thus, the structural mechanism of receptor sharing used by IL-12 family cytokines provides a protein interface blueprint for tuning this cytokine axis for therapeutics.
Collapse
Affiliation(s)
- Caleb R Glassman
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yamuna Kalyani Mathiharan
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin M Jude
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leon Su
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ouliana Panova
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Patrick J Lupardus
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jamie B Spangler
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lauren K Ely
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christoph Thomas
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Georgios Skiniotis
- Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - K Christopher Garcia
- Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Molecular and Cellular Physiology and Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Nguyen KG, Vrabel MR, Mantooth SM, Hopkins JJ, Wagner ES, Gabaldon TA, Zaharoff DA. Localized Interleukin-12 for Cancer Immunotherapy. Front Immunol 2020; 11:575597. [PMID: 33178203 PMCID: PMC7593768 DOI: 10.3389/fimmu.2020.575597] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
Interleukin-12 (IL-12) is a potent, pro-inflammatory type 1 cytokine that has long been studied as a potential immunotherapy for cancer. Unfortunately, IL-12's remarkable antitumor efficacy in preclinical models has yet to be replicated in humans. Early clinical trials in the mid-1990's showed that systemic delivery of IL-12 incurred dose-limiting toxicities. Nevertheless, IL-12's pleiotropic activity, i.e., its ability to engage multiple effector mechanisms and reverse tumor-induced immunosuppression, continues to entice cancer researchers. The development of strategies which maximize IL-12 delivery to the tumor microenvironment while minimizing systemic exposure are of increasing interest. Diverse IL-12 delivery systems, from immunocytokine fusions to polymeric nanoparticles, have demonstrated robust antitumor immunity with reduced adverse events in preclinical studies. Several localized IL-12 delivery approaches have recently reached the clinical stage with several more at the precipice of translation. Taken together, localized delivery systems are supporting an IL-12 renaissance which may finally allow this potent cytokine to fulfill its considerable clinical potential. This review begins with a brief historical account of cytokine monotherapies and describes how IL-12 went from promising new cure to ostracized black sheep following multiple on-study deaths. The bulk of this comprehensive review focuses on developments in diverse localized delivery strategies for IL-12-based cancer immunotherapies. Advantages and limitations of different delivery technologies are highlighted. Finally, perspectives on how IL-12-based immunotherapies may be utilized for widespread clinical application in the very near future are offered.
Collapse
Affiliation(s)
- Khue G Nguyen
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Maura R Vrabel
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Siena M Mantooth
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Jared J Hopkins
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Ethan S Wagner
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Taylor A Gabaldon
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - David A Zaharoff
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
4
|
Kiełbik A, Szlasa W, Saczko J, Kulbacka J. Electroporation-Based Treatments in Urology. Cancers (Basel) 2020; 12:E2208. [PMID: 32784598 PMCID: PMC7465806 DOI: 10.3390/cancers12082208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
The observation that an application of a pulsed electric field (PEF) resulted in an increased permeability of the cell membrane has led to the discovery of the phenomenon called electroporation (EP). Depending on the parameters of the electric current and cell features, electroporation can be either reversible or irreversible. The irreversible electroporation (IRE) found its use in urology as a non-thermal ablative method of prostate and renal cancer. As its mechanism is based on the permeabilization of cell membrane phospholipids, IRE (as well as other treatments based on EP) provides selectivity sparing extracellular proteins and matrix. Reversible EP enables the transfer of genes, drugs, and small exogenous proteins. In clinical practice, reversible EP can locally increase the uptake of cytotoxic drugs such as cisplatin and bleomycin. This approach is known as electrochemotherapy (ECT). Few in vivo and in vitro trials of ECT have been performed on urological cancers. EP provides the possibility of transmission of genes across the cell membrane. As the protocols of gene electrotransfer (GET) over the last few years have improved, EP has become a well-known technique for non-viral cell transfection. GET involves DNA transfection directly to the cancer or the host skin and muscle tissue. Among urological cancers, the GET of several plasmids encoding prostate cancer antigens has been investigated in clinical trials. This review brings into discussion the underlying mechanism of EP and an overview of the latest progress and development perspectives of EP-based treatments in urology.
Collapse
Affiliation(s)
- Aleksander Kiełbik
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (A.K.); (W.S.)
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (A.K.); (W.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| |
Collapse
|
5
|
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
|
6
|
Mukhopadhyay A, Wright J, Shirley S, Canton DA, Burkart C, Connolly RJ, Campbell JS, Pierce RH. Characterization of abscopal effects of intratumoral electroporation-mediated IL-12 gene therapy. Gene Ther 2019; 26:1-15. [PMID: 30323352 PMCID: PMC6514882 DOI: 10.1038/s41434-018-0044-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 12/16/2022]
Abstract
Intratumoral electroporation-mediated IL-12 gene therapy (IT-pIL12/EP) has been shown to be safe and effective in clinical trials, demonstrating systemic antitumor effects with local delivery of this potent cytokine. We recently optimized our IL-12 gene delivery platform to increase transgene expression and efficacy in preclinical models. Here we analyze the immunological changes induced with the new IT-pIL12/EP platform in both electroporated and distant, non-electroporated lesions. IT-pIL12/EP-treated tumors demonstrated rapid induction of IL-12-regulated pathways, as well as other cytokines and chemokines pathways, and upregulation of antigen presentation machinery. The distant tumors showed an increase in infiltrating lymphocytes and gene expression changes indicative of a de novo immune response in these untreated lesions. Flow cytometric analyses revealed a KLRG1hi CD8+ effector T-cell population uniquely present in mice treated with IT-pIL12/EP. Despite being highly activated, this population expressed diminished levels of PD-1 when re-exposed to antigen in the PD-L1-rich tumor. Other T-cell exhaustion markers appeared to be downregulated in concert, suggesting an orchestrated "armoring" of these effector T cells against T-cell checkpoints when primed in the presence of IL-12 in situ. These cells may represent an important mechanism by which local IL-12 gene therapy can induce a systemic antitumor immune response without the associated toxicity of systemic IL-12 exposure.
Collapse
Affiliation(s)
| | - Jocelyn Wright
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
| | - Shawna Shirley
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
| | - David A Canton
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
| | - Christoph Burkart
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
| | - Richard J Connolly
- OncoSec Medical Incorporated, 3565 General Atomics Court #100, San Diego, CA, 92121, USA
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, 98109, USA
| | - Jean S Campbell
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, 98109, USA
| | - Robert H Pierce
- Fred Hutchinson Cancer Center, 1100 Fairview Avenue N, Seattle, WA, 98109, USA.
| |
Collapse
|
7
|
Van der Jeught K, Bialkowski L, Daszkiewicz L, Broos K, Goyvaerts C, Renmans D, Van Lint S, Heirman C, Thielemans K, Breckpot K. Targeting the tumor microenvironment to enhance antitumor immune responses. Oncotarget 2015; 6:1359-81. [PMID: 25682197 PMCID: PMC4359300 DOI: 10.18632/oncotarget.3204] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/24/2014] [Indexed: 12/16/2022] Open
Abstract
The identification of tumor-specific antigens and the immune responses directed against them has instigated the development of therapies to enhance antitumor immune responses. Most of these cancer immunotherapies are administered systemically rather than directly to tumors. Nonetheless, numerous studies have demonstrated that intratumoral therapy is an attractive approach, both for immunization and immunomodulation purposes. Injection, recruitment and/or activation of antigen-presenting cells in the tumor nest have been extensively studied as strategies to cross-prime immune responses. Moreover, delivery of stimulatory cytokines, blockade of inhibitory cytokines and immune checkpoint blockade have been explored to restore immunological fitness at the tumor site. These tumor-targeted therapies have the potential to induce systemic immunity without the toxicity that is often associated with systemic treatments. We review the most promising intratumoral immunotherapies, how these affect systemic antitumor immunity such that disseminated tumor cells are eliminated, and which approaches have been proven successful in animal models and patients.
Collapse
Affiliation(s)
- Kevin Van der Jeught
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lukasz Bialkowski
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lidia Daszkiewicz
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Katrijn Broos
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Dries Renmans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Sandra Van Lint
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| |
Collapse
|
8
|
Tsai WB, Lai HY, Lee JL, Lo CW, Chen WS. Enhancement of the cytotoxicity and selectivity of doxorubicin to hepatoma cells by synergistic combination of galactose-decorated γ-poly(glutamic acid) nanoparticles and low-intensity ultrasound. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:5510-5517. [PMID: 24754730 DOI: 10.1021/la500352g] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Specific drug delivery to solid tumors remains one of the challenges in cancer therapy. The aim of this study was to combine three drug-targeting strategies, polymer-drug conjugate, ligand presentation and ultrasound treatment, to enhance the efficacy and selectivity of doxorubicin (DXR) to hepatoma cells. The conjugation of DXR to γ-poly(glutamic acids) (γ-PGA) decreased the cytotoxicity of DXR, while the conjugation of galactosamine (Gal) to the γ-PGA-DXR conjugate restored the cytotoxic efficacy of DXR on hepatoma cells due to increased uptake of DXR. Furthermore, low-intensity ultrasound treatment increased the cell-killing ability of γ-PGA-DXR conjugates by 20%. The in vitro results showed the potential of the γ-PGA-DXR-Gal conjugate for future clinical applications.
Collapse
Affiliation(s)
- Wei-Bor Tsai
- Department of Chemical Engineering, National Taiwan University , Number 1, Section 4, Roosevelt Road, Taipei 106, Taiwan
| | | | | | | | | |
Collapse
|
9
|
Lee JL, Lo CW, Inserra C, Béra JC, Chen WS. Ultrasound enhanced PEI-mediated gene delivery through increasing the intracellular calcium level and PKC-δ protein expression. Pharm Res 2014; 31:2354-66. [PMID: 24623478 DOI: 10.1007/s11095-014-1332-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/08/2014] [Indexed: 12/16/2022]
Abstract
PURPOSE Polyethylenimine (PEI), a cationic polymer, has been shown to aggregate plasmid DNA and facilitate its internalization. It has also been shown that combining ultrasound (US) with PEI could enhance and prolong in vitro and in vivo transgene expression. However, the role US in the enhancement of PEI uptake is poorly understood. This study investigates the impact of US on PEI-mediated gene transfection. METHODS Specific endocytosis pathway siRNA, including clathrin HC siRNA, caveolin-1 siRNA and protein kinase C-delta (PKC-δ) siRNA, are used to block the corresponding endocytosis pathways prior to the transfection of luciferase DNA/PEI polyplexes to cultured cells by 1-MHz pulsed US with ultrasound contrast agent SonoVue®. RESULTS Transgene expression was found not to be enhanced by US treatment in the presence of the PKC-δ siRNA. We further demonstrated that PKC-δ protein could be enhanced at 6 h after US exposure. Moreover, intracellular calcium levels were found to be significantly increased at 3 h after US exposure, while transgene expressions were significantly reduced in the presence of calcium channel blockers both in vitro and in vivo. CONCLUSIONS Our results suggest that US enhanced PEI-mediated gene transfection specifically by increasing PKC-δ related fluid phase endocytosis, which was induced by increasing the intracellular calcium levels.
Collapse
Affiliation(s)
- Jyun-Lin Lee
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital National Taiwan University College of Medicine, Taipei, Taiwan, Republic of China
| | | | | | | | | |
Collapse
|
10
|
Cao L, Zeng Q, Xu C, Shi S, Zhang Z, Sun X. Enhanced antitumor response mediated by the codelivery of paclitaxel and adenoviral vector expressing IL-12. Mol Pharm 2013; 10:1804-14. [PMID: 23534449 DOI: 10.1021/mp300602j] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
It has been well-established that chemo-immunotherapy using cytotoxic drugs and appropriate cytokines offers a promising approach for the treatment of neoplastic diseases. In view of this, to improve melanoma treatment effect, our study developed a new codelivery system (AL/Ad5/PTX) that paclitaxel (PTX) and adenovirus encoding for murine interleukin-12 (Ad5-mIL-12) were incorporated into anionic liposomes (AL). First, AL/Ad5/PTX complexes were prepared by incorporating Ad5 into anionic PTX liposomes using calcium-induced phase change. Second, the size distribution and zeta potential of AL/Ad5/PTX were investigated. Third, the results of in vitro transduction assays showed that PTX introduced into AL/Ad-luc or AL/Ad5-mIL-12 highly enhanced gene transduction efficiency in B16 cells than naked Ad5 or AL/Ad complexes while it had no comparability in A549 cells. Finally, a melanoma-bearing mouse model was established to assess the antitumor effect. Tumor growth inhibition and prolonged survival time, accompanied by increased mIL-12 or interferon-γ (IFN-γ) expression levels in serum or tumor sites, were observed in mice treated with AL/Ad5-mIL-12/PTX, as compared with those treated with either AL/Ad5-mIL-12 or AL/PTX. In conclusion, these results suggested that codelivery of Ad5-mIL-12 and PTX incorporated into AL could be a relatively efficient strategy for the treatment of melanoma.
Collapse
Affiliation(s)
- Linjie Cao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | | | | | | | | | | |
Collapse
|
11
|
Sin JI, Park JB, Lee IH, Park D, Choi YS, Choe J, Celis E. Intratumoral electroporation of IL-12 cDNA eradicates established melanomas by Trp2(180-188)-specific CD8+ CTLs in a perforin/granzyme-mediated and IFN-γ-dependent manner: application of Trp2(180-188) peptides. Cancer Immunol Immunother 2012; 61:1671-82. [PMID: 22382361 PMCID: PMC11028417 DOI: 10.1007/s00262-012-1214-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 01/27/2012] [Indexed: 01/05/2023]
Abstract
Intratumoral electroporation (IT-EP) with IL-12 cDNA (IT-EP/IL12) can lead to the eradication of established B16 melanoma tumors in mice. Here, we explore the immunological mechanism of the antitumor effects generated by this therapy. The results show that IT-EP/IL12 applied only once resulted in eradication in 70% animals with large established B16 tumors. Tumor eradication required the participation of CD8+ T cells, but not CD4+ T cells and NK cells. IT-EP/IL12 induced antigen-specific CD8+ T cell responses against the immunodominant Trp2(180-188) epitope and generated a systemic response, resulting in significant therapeutic effects against distal, untreated tumors. The therapeutic effect of IT-EP/IL12 was absent in perforin-deficient mice, indicating that tumor elimination occurred through conventional perforin/granzyme lysis by CTLs. Moreover, this therapy induced some degree of immunological memory that protected approximately one-third of the cured mice against a subsequent tumor challenge. Moreover, antitumor efficacy and long-term protection against B16 were significantly improved by concurrent Trp2 peptide immunization through more induction of Ag-specific CTL responses and more attraction of IFN-γ-expressing CD8+ T cells into tumor sites. The antitumor effect of IT-EP/IL12 required the participation of IFN-γ, which was shown to induce MHC class I expression on B16 cells and increase the lytic activity of the CD8+ CTL generated by IT-EP/IL12. The results from these animal studies may help in the development of IT-EP/IL12 for cancer patients.
Collapse
Affiliation(s)
- Jeong-Im Sin
- Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Korea.
| | | | | | | | | | | | | |
Collapse
|
12
|
Nakai N, Kishida T, Hartmann G, Katoh N, Imanishi J, Kishimoto S, Mazda O. Mitf silencing cooperates with IL-12 gene transfer to inhibit melanoma in mice. Int Immunopharmacol 2010; 10:540-5. [PMID: 20074674 DOI: 10.1016/j.intimp.2009.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 11/15/2009] [Accepted: 12/28/2009] [Indexed: 11/15/2022]
Abstract
Malignant melanoma is a malignant neoplasm originating from the melanocyte lineage. Microphthalmia-associated transcription factor (Mitf) is crucially involved in the melanin synthesis as well as proliferation and survival of melanocyte and melanoma. We previously showed that short interfering RNA (siRNA) that is specific for the Mitf gene (Mitf-siRNA) significantly inhibited growth of B16 melanoma after electro-transfected in vivo into preestablished tumor in mice. Here we assessed efficacy of electroporation-mediated co-transfection of Mitf-siRNA and IL-12 gene in the treatment of murine melanoma. As results, the tumor growth was more strongly inhibited by intratumor co-transfection with Mitf-siRNA and IL-12-encoding plasmid DNA than by transfection with either of the molecules alone. The co-transfection induced intratumor infiltration of CD4+ and CD8+ T cells, and hampered neoangiogenesis in the tumor. The findings suggest that the RNAi/cytokine gene combination therapy by means of electroporation may become a novel and efficacious therapeutic modality to treat neoplasms including melanoma.
Collapse
Affiliation(s)
- Noriaki Nakai
- Department of Dermatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamikyo, Kyoto 602-8566, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Aung W, Hasegawa S, Koshikawa-Yano M, Obata T, Ikehira H, Furukawa T, Aoki I, Saga T. Visualization of in vivo electroporation-mediated transgene expression in experimental tumors by optical and magnetic resonance imaging. Gene Ther 2009; 16:830-9. [PMID: 19458649 DOI: 10.1038/gt.2009.55] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Revised: 01/06/2009] [Accepted: 01/06/2009] [Indexed: 11/10/2022]
Abstract
In vivo electroporation (EP) is an efficient method for effective gene transfer and is highly expected for application in anticancer gene therapy. Non-invasive monitoring of gene transfer/expression is critical for optimal gene therapy. Here we report in vivo optical and high-field magnetic resonance imaging (MRI) of EP-mediated transgene expression in a tumor model. Initially, we observed spatio-temporal change in in vivo EP-mediated transgene expression by optical imaging using red fluorescence protein (RFP) as a reporter gene. Next, we constructed a dual-reporter plasmid carrying a gene-encoding MRI reporter ferritin heavy chain and RFP gene to visualize the intratumoral transgene expression by dual modality. Cells transfected with this plasmid showed lower signal intensity on in vitro T(2)-weighted cellular MRI and quantitatively increased the transverse relaxation rate (1/T(2)) compared with control cells. After conducting in vivo EP in an experimental tumor, the plasmid-injected region showed both fluorescent emissions in optical imaging and detectably lowered signal on T(2)-weighted MRI. The correlative immunohistological findings confirmed that both the reporter transgenes were co-expressed in this region. Thus, our strategy provides a platform for evaluating EP-mediated cancer gene therapy easily and safely without administering contrast agent or substrate.
Collapse
Affiliation(s)
- W Aung
- Diagnostic Imaging Group, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Henshaw JW, Yuan F. Field distribution and DNA transport in solid tumors during electric field-mediated gene delivery. J Pharm Sci 2008; 97:691-711. [PMID: 17624918 DOI: 10.1002/jps.21000] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gene therapy has a great potential in cancer treatment. However, the efficacy of cancer gene therapy is currently limited by the lack of a safe and efficient means to deliver therapeutic genes into the nucleus of tumor cells. One method under investigation for improving local gene delivery is based on the use of pulsed electric field. Despite repeated demonstration of its effectiveness in vivo, the underlying mechanisms behind electric field-mediated gene delivery remain largely unknown. Without a thorough understanding of these mechanisms, it will be difficult to further advance the gene delivery. In this review, the electric field-mediated gene delivery in solid tumors will be examined by following individual transport processes that must occur in vivo for a successful gene transfer. The topics of examination include: (i) major barriers for gene delivery in the body, (ii) distribution of electric fields at both cell and tissue levels during the application of external fields, and (iii) electric field-induced transport of genes across each of the barriers. Through this approach, the review summarizes what is known about the mechanisms behind electric field-mediated gene delivery and what require further investigations in future studies.
Collapse
Affiliation(s)
- Joshua W Henshaw
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, Durham, North Carolina 27708, USA
| | | |
Collapse
|
15
|
Horton HM, Lalor PA, Rolland AP. IL-2 plasmid electroporation: from preclinical studies to phase I clinical trial. Methods Mol Biol 2008; 423:361-372. [PMID: 18370214 DOI: 10.1007/978-1-59745-194-9_28] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Electroporation (EP)-assisted intralesional delivery of Interleukin-2 (IL-2) plasmid (pDNA) has the potential to increase the local concentration of the expressed cytokine for an extended time in the injected tumors while minimizing its systemic concentration, in comparison with systemic delivery of the recombinant cytokine. Nonclinical Investigational New Drug application-enabling studies were performed in mice to evaluate the effect of intratumoral administration of murine IL-2 pDNA on local expression and systemic distribution of IL-2 transgene as well as the inhibition of established tumor growth. The safety of repeated administrations of a human IL-2 pDNA product candidate with EP was evaluated in rats. Following the nonclinical safety and efficacy studies, a human IL-2 pDNA product candidate intralesionally administered with EP to metastatic melanoma patients is currently being investigated in a phase I clinical trial.
Collapse
|
16
|
Abstract
The importance of DNA formulation in safe and efficient electrogene transfer is increasingly recognized as electroporation technology enters into clinical development. A phenomenal increase in naked DNA delivery by electroporation offers new opportunities for nonviral gene therapies previously considered difficult because of insufficient delivery. However, significant tissue damage related to harsh electroporation conditions raises serious safety concerns with the use of electroporation in healthy tissues, which limits its current applications to only nonhealthy tissues such as tumors. DNA formulations designed to minimize tissue damage or enhance expression at weaker electric pulses have been examined to address these concerns. These include formulations fortified with the addition of transfection reagent(s), membrane-permeating agents, tissue matrix modifiers, targeted ligands, or agents modifying electrical conductivity or membrane stability to enhance delivery efficiency or reduce tissue damage. These advancements in DNA formulation could prove to be useful in improving the safety of electroporation protocols for human applications.
Collapse
|
17
|
Lim DS, Kim JH, Lee DS, Yoon CH, Bae YS. DC immunotherapy is highly effective for the inhibition of tumor metastasis or recurrence, although it is not efficient for the eradication of established solid tumors. Cancer Immunol Immunother 2007; 56:1817-29. [PMID: 17443323 PMCID: PMC11029899 DOI: 10.1007/s00262-007-0325-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2006] [Accepted: 03/23/2007] [Indexed: 12/21/2022]
Abstract
Dendritic cell (DC)-based immunotherapy has not been as effective as expected in most solid tumors even in the murine model, particularly in renal cell carcinoma (RCC). Our investigation was initiated to identify what causes the limitations of DC-based immunotherapy in solid RCC. We have investigated immunosuppressive factors from tumors and their effects on DC migration, as well as cytotoxic T lymphocyte (CTL) response and lymphocyte infiltration into the tumor mass upon vaccination with mouse renal adenocarcinoma (Renca) cell lysate-pulsed bone marrow (Bm)-derived DC in tumor-bearing mice. We also investigated pulmonary metastasis- and tumor recurrence-inhibitory effects of DC-vaccination in the solid tumor-bearing mice. In these experiments, we found that the limitations of DC-based immunotherapy to solid RCC likely result from tumor-mediated TGF-beta hindrance of immune attack rather than insufficient immune induction by DC therapy. In fact, the CTL response induced by DC therapy was quite sufficient and functional for the inhibition of tumor recurrence after surgery or of tumor metastasis induced by additional tumor-challenge to the tumor-bearing mice. Taken together, our present results obtained in mouse model suggest the potential of DC immunotherapy in tumor patients for hindering or blocking disease progression by inhibition of tumor metastasis and/or tumor recurrence after surgery.
Collapse
Affiliation(s)
- Dae-Seog Lim
- Research Institute for DC immunotherapy, CreaGene Inc., 2F Jungang Induspia V, 138-6 Sangdaewon-dong, Jungwon-gu, Seongnam-si, Gyeonggi-do, 462-120 South Korea
| | - Jeong-Hwan Kim
- Research Institute for DC immunotherapy, CreaGene Inc., 2F Jungang Induspia V, 138-6 Sangdaewon-dong, Jungwon-gu, Seongnam-si, Gyeonggi-do, 462-120 South Korea
| | - Dong-Seong Lee
- Research Institute for DC immunotherapy, CreaGene Inc., 2F Jungang Induspia V, 138-6 Sangdaewon-dong, Jungwon-gu, Seongnam-si, Gyeonggi-do, 462-120 South Korea
| | - Cheol-Hee Yoon
- Department of Biological Science, Sungkyunkwan University, 300 Cheoncheon-dong, Suwon, Gyeonggi, 440–746 South Korea
| | - Yong-Soo Bae
- Research Institute for DC immunotherapy, CreaGene Inc., 2F Jungang Induspia V, 138-6 Sangdaewon-dong, Jungwon-gu, Seongnam-si, Gyeonggi-do, 462-120 South Korea
- Department of Biological Science, Sungkyunkwan University, 300 Cheoncheon-dong, Suwon, Gyeonggi, 440–746 South Korea
| |
Collapse
|
18
|
Henshaw JW, Zaharoff DA, Mossop BJ, Yuan F. Electric field-mediated transport of plasmid DNA in tumor interstitium in vivo. Bioelectrochemistry 2007; 71:233-42. [PMID: 17728192 PMCID: PMC2885976 DOI: 10.1016/j.bioelechem.2007.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Revised: 07/07/2007] [Accepted: 07/18/2007] [Indexed: 12/11/2022]
Abstract
Local pulsed electric field application is a method for improving non-viral gene delivery. Mechanisms of the improvement include electroporation and electrophoresis. To understand how electrophoresis affects pDNA delivery in vivo, we quantified the magnitude of electric field-induced interstitial transport of pDNA in 4T1 and B16.F10 tumors implanted in mouse dorsal skin-fold chambers. Four different electric pulse sequences were used in this study, each consisted of 10 identical pulses that were 100 or 400 V/cm in strength and 20 or 50 ms in duration. The interval between consecutive pulses was 1 s. The largest distance of transport was obtained with the 400 V/cm and 50 ms pulse, and was 0.23 and 0.22 microm/pulse in 4T1 and B16.F10 tumors, respectively. There were no significant differences in transport distances between 4T1 and B16.F10 tumors. Results from in vivo mapping and numerical simulations revealed an approximately uniform intratumoral electric field that was predominantly in the direction of the applied field. The data in the study suggested that interstitial transport of pDNA induced by a sequence of ten electric pulses was ineffective for macroscopic delivery of genes in tumors. However, the induced transport was more efficient than passive diffusion.
Collapse
Affiliation(s)
| | | | | | - Fan Yuan
- Corresponding author: Dr. Fan Yuan Department of Biomedical Engineering Duke University 136 Hudson Hall Durham, NC 27708 (919) 660 – 5411 (phone) (919) 684 – 4488 (fax)
| |
Collapse
|
19
|
Dobaño C, Widera G, Rabussay D, Doolan DL. Enhancement of antibody and cellular immune responses to malaria DNA vaccines by in vivo electroporation. Vaccine 2007; 25:6635-45. [PMID: 17669562 DOI: 10.1016/j.vaccine.2007.06.036] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 05/08/2007] [Accepted: 06/13/2007] [Indexed: 01/16/2023]
Abstract
We evaluated the effectiveness of in vivo electroporation (EP) for the enhancement of immune responses induced by DNA plasmids encoding the pre-erythrocytic Plasmodium yoelii antigens PyCSP and PyHEP17 administered intramuscularly and intradermally to mice. EP resulted in a 16- and 2-fold enhancement of antibody responses to PyCSP and PyHEP17, respectively. Immunization with 5 microg of DNA via EP was equivalent to 50 microg of DNA via conventional needle, thus reducing by 10-fold the required dose to produce a given effect. Moreover, IFN-gamma responses were increased by approximately 2-fold. Data demonstrate the potential of EP to enhance immune responses to DNA vaccines against infectious agents.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan/immunology
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Electroporation/methods
- Female
- Immunity, Cellular/immunology
- Immunization/methods
- Injections, Intradermal
- Injections, Intramuscular
- Interferon-gamma/immunology
- Liver/immunology
- Liver/parasitology
- Malaria Vaccines/administration & dosage
- Malaria Vaccines/immunology
- Mice
- Mice, Inbred BALB C
- Plasmids/genetics
- Plasmodium yoelii/genetics
- Plasmodium yoelii/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Carlota Dobaño
- Malaria Program, Naval Medical Research Center, Silver Spring, MD 20910-7500, United States.
| | | | | | | |
Collapse
|
20
|
Cloning and characterization of an adenoviral vector for highly efficient and doxycycline-suppressible expression of bioactive human single-chain interleukin 12 in colon cancer. BMC Biotechnol 2007; 7:35. [PMID: 17594499 PMCID: PMC1913502 DOI: 10.1186/1472-6750-7-35] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2007] [Accepted: 06/26/2007] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Interleukin-12 (IL-12) is well characterized to induce cellular antitumoral immunity by activation of NK-cells and T-lymphocytes. However, systemic administration of recombinant human IL-12 resulted in severe toxicity without perceptible therapeutic benefit. Even though intratumoral expression of IL-12 leads to tumor regression and long-term survival in a variety of animal models, clinical trials have not yet shown a significant therapeutic benefit. One major obstacle in the treatment with IL-12 is to overcome the relatively low expression of the therapeutic gene without compromising the safety of such an approach. Our objective was to generate an adenoviral vector system enabling the regulated expression of very high levels of bioactive, human IL-12. RESULTS High gene expression was obtained utilizing the VP16 herpes simplex transactivator. Strong regulation of gene expression was realized by fusion of the VP16 to a tetracycline repressor with binding of the fusion protein to a flanking tetracycline operator and further enhanced by auto-regulated expression of its fusion gene within a bicistronic promoter construct. Infection of human colon cancer cells (HT29) at a multiplicity of infection (m.o.i.) of 10 resulted in the production of up to 8000 ng/106 cells in 48 h, thus exceeding any published vector system so far. Doxycycline concentrations as low as 30 ng/ml resulted in up to 5000-fold suppression, enabling significant reduction of gene expression in a possible clinical setting. Bioactivity of the human single-chain IL-12 was similar to purified human heterodimeric IL-12. Frozen sections of human colon cancer showed high expression of the coxsackie adenovirus receptor with significant production of human single chain IL-12 in colon cancer biopsies after infection with 3*107 p.f.u. Ad.3r-scIL12. Doxycycline mediated suppression of gene expression was up to 9000-fold in the infected colon cancer tissue. CONCLUSION VP16 transactivator-mediated and doxycycline-regulated expression of the human interleukin-12 gene enables highly efficient and tightly controlled cytokine expression in human cancer. These data illustrate the potential of the described adenoviral vector system for the safe and superior expression of therapeutic genes in the treatment of colorectal cancer and other malignancies.
Collapse
|
21
|
Affiliation(s)
- Loree C Heller
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | | |
Collapse
|
22
|
Abstract
The utilisation of nonviral gene delivery methods has been increasing steadily, however, a drawback has been the relative low efficiency of gene transfer with naked DNA compared with viral delivery methods. In vivo electroporation, which has previously been used clinically to deliver chemotherapeutic agents, also enhances the delivery of plasmid DNA and has been used to deliver plasmids to several tissue types, particularly muscle and tumour. Recently, a large number of preclinical studies for a variety of therapeutic modalities have demonstrated the potential of electrically mediated gene transfer. Although clinical trials using gene transfer with in vivo electroporation have not as yet been realised, the tremendous growth of this technology suggests that the first trials will soon be initiated.
Collapse
MESH Headings
- Animals
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/therapy
- Electroporation
- Erythropoietin/administration & dosage
- Erythropoietin/genetics
- Erythropoietin/metabolism
- Forecasting
- Gene Expression Regulation
- Gene Transfer Techniques
- Genetic Therapy/methods
- Genetic Therapy/trends
- Growth Substances/administration & dosage
- Growth Substances/genetics
- Growth Substances/metabolism
- Hematologic Diseases/genetics
- Hematologic Diseases/metabolism
- Hematologic Diseases/therapy
- Humans
- Injections, Intramuscular
- Interleukin-12/administration & dosage
- Interleukin-12/genetics
- Interleukin-12/metabolism
- Melanoma/genetics
- Melanoma/metabolism
- Melanoma/therapy
- Plasmids/administration & dosage
- Plasmids/genetics
- Plasmids/metabolism
- Protein Deficiency/genetics
- Protein Deficiency/metabolism
- Protein Deficiency/therapy
- Toxins, Biological/administration & dosage
- Toxins, Biological/genetics
- Toxins, Biological/metabolism
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/metabolism
Collapse
Affiliation(s)
- Loree C Heller
- University of South Florida, Center of Molecular Delivery, Department of Medical Microbiology and Immunology, Center for Molecular Delivery, College of Medicine, Tampa, 33612-4799, USA.
| | | | | |
Collapse
|
23
|
Salem ML, Gillanders WE, Kadima AN, El-Naggar S, Rubinstein MP, Demcheva M, Vournakis JN, Cole DJ. Review: novel nonviral delivery approaches for interleukin-12 protein and gene systems: curbing toxicity and enhancing adjuvant activity. J Interferon Cytokine Res 2006; 26:593-608. [PMID: 16978064 DOI: 10.1089/jir.2006.26.593] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
It has become increasingly apparent that the ability to generate an optimal host immune response requires effective cross talk between the innate and adaptive components of the immune system. Pro-inflammatory cytokines, in particular those that can induce a danger signal, often called signal 3, are crucial in this role of initiating and augmenting the presentation of exogenous antigen to T cells by dendritic cells. Interleukin-12 (IL-12) in particular has been defined as a "signal 3" cytokine required for the antigen cross priming. Given this unique interactive function, a significant amount of work has been performed to define possible therapeutic applications for IL-12. Systemic IL-12 administration can clearly act as a potent adjuvant for postvaccination T cell responses in a variety of diseases. As an example, in the cancer setting, systemic IL-12 is capable of suppressing tumor growth, metastasis, and angiogenesis in vivo. IL-12, however, has been associated with significant dose- and schedule-dependent toxicity in early clinical trials, results that have proven to be a major obstacle to its clinical application. Recent research has focused on decreasing the toxicity of IL-12 using different delivery approaches, including virus-based and gene-modified cell-based delivery. Although effective, these approaches also have limitations, including the generation of neutralizing antibodies, in addition to lacking the simplicity and versatility required for universal clinical application. Thus, there is a significant interest in the development of alternative delivery approaches for IL-12 administration that can overcome these issues. Several nonviral delivery approaches for IL-12 protein or gene expression vectors are being defined, including alum, liposomes, and polymer-based delivery. These developing approaches have shown promising adjuvant effects with significantly lessened systemic toxicity. This article discusses the potential capabilities of these nonvirus-based IL-12 delivery systems in different disease settings, including allergy, infection, and cancer.
Collapse
Affiliation(s)
- Mohamed Labib Salem
- Department of Surgery, Section of Surgical Oncology, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
McCray AN, Ugen KE, Muthumani K, Kim JJ, Weiner DB, Heller R. Complete regression of established subcutaneous B16 murine melanoma tumors after delivery of an HIV-1 Vpr-expressing plasmid by in vivo electroporation. Mol Ther 2006; 14:647-55. [PMID: 16950655 DOI: 10.1016/j.ymthe.2006.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 06/06/2006] [Accepted: 06/13/2006] [Indexed: 11/25/2022] Open
Abstract
Novel therapies and delivery methods directed against malignancies such as melanoma, and particularly metastatic melanoma, are needed. The HIV-1 accessory protein Vpr (viral protein R) has previously been demonstrated to induce G2 cell cycle arrest as well as in vitro growth inhibition/killing of a number of tumor cells by apoptosis. In vivo electroporation has been utilized as an effective delivery method for pharmacologic agents and DNA plasmids that express "therapeutic" proteins and has been targeted to various tissues, including malignant tumors. For the study reported here, we hypothesized that intratumoral delivery of a Vpr expression plasmid through in vivo electroporation would induce apoptosis and growth attenuation or regression of melanoma tumors. Established subcutaneous B16.F10 melanoma tumors were injected intratumorally with a Vpr-expressing (either 25 or 100 microg) plasmid, followed by electroporation, on day 0 (i.e., when tumors had attained an appropriate size) and day 4. Treatment with 25 or 100 microg of the Vpr-expressing plasmid resulted in complete tumor regression with long-term survival in 14.3 and 7.1% of the mice, respectively. In addition, electroporative delivery of the Vpr-expressing plasmid was shown to induce apoptosis in tumors after intratumoral injection. This is the first report demonstrating the ability of Vpr, when delivered as a DNA expression plasmid with in vivo electroporation, to attenuate melanoma lesion growth and induce complete tumor regression coupled with long-term survival of mice in a highly aggressive and metastatic solid tumor model.
Collapse
Affiliation(s)
- Andrea N McCray
- Department of Medical Microbiology and Immunology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | | | | | | | | | | |
Collapse
|
25
|
|
26
|
Lavigne MD, Górecki DC. Emerging vectors and targeting methods for nonviral gene therapy. Expert Opin Emerg Drugs 2006; 11:541-57. [PMID: 16939390 DOI: 10.1517/14728214.11.3.541] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Until recently, nonviral vectors were outside the mainstream of gene transfer technology. Recent problems in clinical trials using viral vectors renewed interest in these methods. The clinical usefulness of nonviral methods is still hindered by their relatively low gene delivery/transgene expression efficiencies. Vectors must navigate a series of obstacles before the therapeutic gene can be expressed. This review considers these barriers and the properties of components of nonviral vectors that are essential for nucleic acid transfer. Although developments of new physical methods (hydrodynamic delivery, ultrasound, electroporation) have made a significant impact on gene transfer efficiency, various chemical carriers (lipids and polymers) have been shown to achieve high-level gene delivery and functional expression. Success of nonviral gene targeting will depend not only on the efficacy, but also safety of this methodology, and this aspect is also discussed. Understanding problems associated with nonviral targeting can also help in designing better viral vectors. In fact, interplay between viral and nonviral technologies should lead to a continued refinement of both methodologies.
Collapse
Affiliation(s)
- Matthieu D Lavigne
- University of Portsmouth, School of Pharmacy and Biomedical Sciences, St. Michael's Building, White Swan Road, Portsmouth, UK
| | | |
Collapse
|
27
|
Peng B, Zhao Y, Lu H, Pang W, Xu Y. In vivo plasmid DNA electroporation resulted in transfection of satellite cells and lasting transgene expression in regenerated muscle fibers. Biochem Biophys Res Commun 2005; 338:1490-8. [PMID: 16271701 DOI: 10.1016/j.bbrc.2005.10.111] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Accepted: 10/13/2005] [Indexed: 11/24/2022]
Abstract
In vivo plasmid DNA electroporation resulted in elevated and lasting transgene expression in skeletal muscles. But the nature of the cells that contributed to sustained gene expression remains unknown. We followed the fate of plasmid DNA delivered with electroporation and systematically investigated the time course and location of transgene expression in muscle tissues both with GFP and luciferase. Furthermore, satellite cell activation after electroporation was confirmed by RT-PCR and immunohistochemistry analysis. The activated satellite cells were shown to be able to uptake the injected plasmid DNA and express transgene products as regenerated myocytes. We found that cells with longer gene expression durations were mostly regenerated muscle fibers. In contrast, expression in pre-existing muscle fibers was rather transient. We also presented in this study that immune response to transgene products might hamper the lasting gene expression. Based on these observations, we proposed that the underlying mechanism for prolonged transgene expression in the muscles after electroporation is related to the activation and transfection of myogenic satellite cells which subsequently developed into regenerated muscle fibers.
Collapse
Affiliation(s)
- Baowei Peng
- School of Life Sciences and Biotechnology, Shanghai Jiaotong University, Shanghai, PR China
| | | | | | | | | |
Collapse
|
28
|
Mir LM, Moller PH, André F, Gehl J. Electric pulse-mediated gene delivery to various animal tissues. ADVANCES IN GENETICS 2005; 54:83-114. [PMID: 16096009 DOI: 10.1016/s0065-2660(05)54005-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Electroporation designates the use of electric pulses to transiently permeabilize the cell membrane. It has been shown that DNA can be transferred to cells through a combined effect of electric pulses causing (1) permeabilization of the cell membrane and (2) an electrophoretic effect on DNA, leading the polyanionic molecule to move toward or across the destabilized membrane. This process is now referred to as DNA electrotransfer or electro gene transfer (EGT). Several studies have shown that EGT can be highly efficient, with low variability both in vitro and in vivo. Furthermore, the area transfected is restricted by the placement of the electrodes, and is thus highly controllable. This has led to an increasing use of the technology to transfer reporter or therapeutic genes to various tissues, as evidenced from the large amount of data accumulated on this new approach for non-viral gene therapy, termed electrogenetherapy (EGT as well). By transfecting cells with a long lifetime, such as muscle fibers, a very long-term expression of genes can be obtained. A great variety of tissues have been transfected successfully, from muscle as the most extensively used, to both soft (e.g., spleen) and hard tissue (e.g., cartilage). It has been shown that therapeutic levels of systemically circulating proteins can be obtained, opening possibilities for using EGT therapeutically. This chapter describes the various aspects of in vivo gene delivery by means of electric pulses, from important issues in methodology to updated results concerning the electrotransfer of reporter and therapeutic genes to different tissues.
Collapse
Affiliation(s)
- Lluis M Mir
- Laboratory of Vectorology and Gene Transfer, UMR 8121 CNRS Institut Gustave-Roussy, F-94805 Villejuif Cédex, France
| | | | | | | |
Collapse
|
29
|
André F, Mir LM. DNA electrotransfer: its principles and an updated review of its therapeutic applications. Gene Ther 2004; 11 Suppl 1:S33-42. [PMID: 15454955 DOI: 10.1038/sj.gt.3302367] [Citation(s) in RCA: 181] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The use of electric pulses to transfect all types of cells is well known and regularly used in vitro for bacteria and eukaryotic cells transformation. Electric pulses can also be delivered in vivo either transcutaneously or with electrodes in direct contact with the tissues. After injection of naked DNA in a tissue, appropriate local electric pulses can result in a very high expression of the transferred genes. This manuscript describes the evolution in the concepts and the various optimization steps that have led to the use of combinations of pulses that fit with the known roles of the electric pulses in DNA electrotransfer, namely cell electropermeabilization and DNA electrophoresis. A summary of the main applications published until now is also reported, restricted to the in vivo preclinical trials using therapeutic genes.
Collapse
Affiliation(s)
- F André
- Laboratory of Vectorology and Gene Transfer, UMR 8121 CNRS - Institut Gustave-Roussy, Villejuif Cedex, France
| | | |
Collapse
|
30
|
Goto T, Nishi T, Kobayashi O, Tamura T, Dev SB, Takeshima H, Kochi M, Kuratsu JI, Sakata T, Ushio Y. Combination electro-gene therapy using herpes virus thymidine kinase and interleukin-12 expression plasmids is highly efficient against murine carcinomas in vivo. Mol Ther 2004; 10:929-37. [PMID: 15509510 DOI: 10.1016/j.ymthe.2004.07.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Accepted: 07/27/2004] [Indexed: 10/26/2022] Open
Abstract
We report the use of plasmid DNA-mediated combination gene therapy for tumor-bearing mice using in vivo electroporation, also called electro-gene therapy (EGT), that resulted in uncomplicated and complete cures in more than 90% of the mice. Subcutaneously inoculated CT26 tumors in syngeneic BALB/c mice were subjected to repeated EGT treatments consisting of intratumoral co-injection of naked plasmids encoding the cytokine interleukin-12 (IL-12) (p35 and p40 subunits) and the suicide gene herpes simplex virus thymidine kinase (HSV-tk), followed by in vivo electroporation. The early anti-tumor effect was always stronger, and the rate of cure, as seen in the long-term follow-up, was always greater in the groups treated with combination EGT than in those treated with IL-12 or HSV-tk EGT alone. Systemic levels of IL-12 and IFN-gamma increased in both combination and IL-12-alone EGT-treated groups. Moreover, combination EGT for established subcutaneous tumors strongly reduced hematogenous lung metastases and increased survival time when live CT26 tumor cells were injected through the tail vein. Limited experiments on C57/B16 mice with murine melanoma also showed very similar trends. These results suggest that this simple and safe method of plasmid-mediated combination EGT may provide a potentially effective gene therapy for cancer.
Collapse
Affiliation(s)
- Tomoaki Goto
- Department of Neurosurgery, Saiseikai Kumamoto Hospital, 5-3-1 Chikami, Kumamoto 861-4193, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kawai M, Bessho K, Kaihara S, Sonobe J, Oda K, Iizuka T, Maruyama H. Ectopic bone formation by human bone morphogenetic protein-2 gene transfer to skeletal muscle using transcutaneous electroporation. Hum Gene Ther 2004; 14:1547-56. [PMID: 14577916 DOI: 10.1089/104303403322495052] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Therapy using recombinant human bone morphogenetic protein-2 (rhBMP-2) is expected to promote bone healing and regeneration. Previous studies using protein or virus vectors for direct clinical application had problems, including a lack of efficiency, safety, and simplicity of the delivery system, and required an expensive protein, carrier matrix, or antigenic viral vector. In vivo gene transfer by electroporation is a simple and inexpensive method that only requires a plasmid and an electroporation device. Here, we created a plasmid-based human BMP-2 construct (pCAGGS-BMP-2) and examined the induction of bone in the skeletal muscle of rats after transferring different doses of this plasmid (25 microg, 100 microg, and 400 microg) by transcutaneous electroporation (8 electrical pulses of 100 V and 50 msec, in 1 to 5 sessions). First, we verified the gene transfer by transcutaneous electroporation using pCAGGS-lacZ. Next, the BMP-2 gene transfer and the production and localization of BMP-2 were identified by reverse transcription-polymerase chain reaction (RT-PCR), Western blots, and immunohistochemistry. Ectopic bone formation was verified by radiography, histologic and immunohistochemical analyses, and quantitative examination. Ectopic bone formation, consisting of active osteoblasts and osteoclasts, was observed in all rats treated with electroporation. Thus, transcutaneous electroporation with pCAGGS-BMP-2 induced ectopic bone formation in the skeletal muscle of rats. This supports the possibility of applying human BMP-2 gene transfer using transcutaneous electroporation clinically.
Collapse
Affiliation(s)
- Mariko Kawai
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Bloquel C, Bessis N, Boissier MC, Scherman D, Bigey P. Gene Therapy of Collagen-Induced Arthritis by Electrotransfer of Human Tumor Necrosis Factor-αSoluble Receptor I Variants. Hum Gene Ther 2004; 15:189-201. [PMID: 14975191 DOI: 10.1089/104303404772679995] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Electrotransfer is a simple and efficient strategy of nonviral gene delivery. We have used this method to deliver plasmids encoding three human tumor necrosis factor-alpha soluble receptor I variants (hTNFR-Is) a monomeric hTNFR-Is, a chimeric hTNFR-Is/mIgG1, and a dimeric (hTNFR-Is)(2) form. Electrotransfer parameters were studied and because anti-TNF strategies have proven efficient for the treatment of rheumatoid arthritis in clinics, we used a collagen-induced arthritis (CIA) mouse model to assess the efficacy of our constructs in the treatment of the disease. All proteins were proven bioactive, both in vitro and ex vivo. Plasmid intramuscular electrotransfer in mice resulted in a local expression of the three variants for at least 6 months; systemic expression lasted also more than 6 months for the hTNFR-Is/mIgG1 form, while it was shorter for the two other forms. This expression was plasmid dose-dependent. Electrotransfer of 50 microg of hTNFR-Is/mIgG1 at the onset of a CIA induced a clear-cut decrease in both clinical and histologic signs of the disease; the dimeric form also showed some efficacy. Moreover, the long-lasting protective effect was observed for more than 5 weeks. Comparison of this electrotransfer approach with repeated recombinant protein (etanercept) injections highlighted the potential practical interest of gene therapy approach for CIA, which leads to sustained therapeutic effect after single treatment. These results show that electrotransfer may be a useful method to deliver cytokine or anticytokine therapy in rheumatoid arthritis and also illustrate the potentiality of plasmid intramuscular electrotransfer for the rapid screening and assessment of different variant forms of secreted proteins.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/blood
- Antigens, CD/genetics
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/pathology
- Arthritis, Experimental/therapy
- Base Sequence
- Collagen
- Dimerization
- Dose-Response Relationship, Drug
- Electrochemistry
- Electroporation/methods
- Gene Transfer Techniques
- Genetic Therapy/methods
- Genetic Vectors
- Humans
- Injections, Intramuscular
- Mice
- Molecular Sequence Data
- Plasmids/genetics
- Plasmids/metabolism
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/blood
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type I
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Carole Bloquel
- UPRES EA-3408 and Service de Rhumatologie, UFR Léonard de Vinci, Université Paris 13 and Hopital Avicenne (AP-HP), 93017 Bobigny Cedex, France
| | | | | | | | | |
Collapse
|
33
|
Kishida T, Asada H, Itokawa Y, Yasutomi K, Shin-Ya M, Gojo S, Cui FD, Ueda Y, Yamagishi H, Imanishi J, Mazda O. Electrochemo-gene therapy of cancer: intratumoral delivery of interleukin-12 gene and bleomycin synergistically induced therapeutic immunity and suppressed subcutaneous and metastatic melanomas in mice. Mol Ther 2003; 8:738-45. [PMID: 14599806 DOI: 10.1016/j.ymthe.2003.08.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To treat established melanoma in mice, intratumoral transfer of bleomycin and/or an interleukin (IL)-12 expression vector was performed by means of electroporation. Although either bleomycin alone or the IL12 gene alone significantly suppressed the subcutaneous tumors, the combination therapy drastically improved the therapeutic outcome. Three of eight mice (37.5%) that received both bleomycin and the IL12 gene showed complete remission of the preestablished tumors and rejected subsequent rechallenge with the tumor cells. We also examined whether electrochemo-gene therapy for subcutaneous tumor mass induced suppression of pulmonary metastasis that had been established by intravenous inoculation of the melanoma cells. Although metastatic foci were significantly reduced in number in groups that were given IL12 gene alone or bleomycin plus IL12 gene, it was only the combination therapy that significantly prolonged the mean survival period of the tumor-bearing animals. Natural killer (NK) and cytotoxic T lymphocyte cytolytic activities were markedly enhanced in the mice that received the chemo-gene therapy, while IL12 gene therapy alone partially elevated the NK cytotoxicity. The present study suggests that the electroporation-mediated delivery of the IL12 gene and bleomycin synergistically elicits innate and adaptive anti-melanoma immune responses, resulting in marked suppression of the treated tumors as well as bystander metastatic lesions.
Collapse
Affiliation(s)
- Tsunao Kishida
- Department of Microbiology, Kyoto Prefectural University of Medicine, Kamikyo, 602-8566, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Zheng S, Xiao ZX, Pan YL, Han MY, Dong Q. Continuous release of interleukin 12 from microencapsulated engineered cells for colon cancer therapy. World J Gastroenterol 2003; 9:951-5. [PMID: 12717836 PMCID: PMC4611403 DOI: 10.3748/wjg.v9.i5.951] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the anti-tumor immunity against CT26 colon tumor of the microencapsulated cells modified with murine interleukine-12 (mIL-12) gene.
METHODS: Mouse fibroblasts (NIH3T3) were stably transfected to express mIL-12 using expression plasmids carrying mIL-12 gene (p35 and p40), and NIH3T3-mIL-12 cells were encapsulated in alginate microcapsules for long-term delivery of mIL-12. mIL-12 released from the microencapsulated NIH3T3-mIL-12 cells was confirmed using ELISA assay. Transplantation of the microencapsulated NIH3T3-mIL-12 cells was performed in the tumor-bearing mice with CT26 cells. The anti-tumor responses and the anti-tumor activities of the microencapsulated NIH3T3-mIL-12 cells were evaluated.
RESULTS: Microencapsulated NIH3T3-mIL-12 cells could release mIL-12 continuously and stably for a long time. After the microencapsulated NIH3T3-mIL-12 cells were transplanted subcutaneously into the tumor-bearing mice for 21 d, the serum concentrations of mIL-12, mIL-2 and mIFN-γ, the cytotoxicity of the CTL from the splenocytes and the NK activity in the treatment group were significantly higher than those in the controls. Moreover, mIL-12 released from the microencapsulated NIH3T3-mIL-12 cells resulted in a significant inhibition of tumor proliferation and a prolonged survival of tumor-bearing mice.
CONCLUSION: The microencapsulated NIH3T3-mIL-12 cells have a significant therapeutic effect on the experimental colon tumor by activating anti-tumor immune responses in vivo. Microencapsulated and genetically engineered cells may be an extremely versatile tool for tumor gene therapy.
Collapse
Affiliation(s)
- Shu Zheng
- Cancer Institute, Zhejiang University, Hangzhou 310009, Zhejiang Province, China.
| | | | | | | | | |
Collapse
|
35
|
Abstract
The success of gene therapy is largely dependent on the development of the gene delivery vector. Recently, gene transfection into target cells using naked DNA, which is a simple and safe approach, has been improved by combining several physical techniques, for example, electroporation, gene gun, ultrasound and hydrodynamic pressure. Chemical approaches have been utilized to improve the efficiency and cell specificity of gene transfer. Novel gene carrier molecules, which facilitate DNA escape from the endosome into the cytosol, have been developed. Several functional polymers, which enable controlled release of DNA in response to an environmental change, have also been reported. Plasmids with reduced number of CpG motifs, the use of PCR fragments and the sequential injection method have been established for the reduction of immune response triggered by plasmid DNA. Construction of a long-lasting gene expression system is also an important theme for nonviral gene therapy. To date, tissue-specific expression, self-replicating and integrating plasmid systems have been reported. Improvement of delivery methods together with intelligent design of the DNA itself has brought about large degrees of enhancement in the efficiency, specificity and temporal control of nonviral vectors.
Collapse
Affiliation(s)
- T Niidome
- Center for Pharmacogenetics, School of Pharmacy, 633 Salk Hall, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
36
|
Abstract
In vivo electrotransfer is a physical technique for gene delivery in various mammalian tissues, which involves the injection of plasmid DNA into a target tissue and administration of an electric field. Its ease of performance, as well as recent understanding of its mechanism and applications to different mammalian tissues such as skeletal muscle, liver, brain and tumors, makes it a powerful technique. It could be used in gene therapy and as a laboratory tool to study gene functions.
Collapse
Affiliation(s)
- Pascal Bigey
- Laboratoire de Chimie Bioorganique et de Biotechnologie Moléculaire et Cellulaire, UMR 7001 ENSCP/CNRS/Aventis, CRVA-Aventis, Batiment Monod, Laboratory 3C05, 13 quai Jules Guesde, 94403 Vitry-sur-Seine cedex, France
| | | | | |
Collapse
|
37
|
Abstract
The use of electric pulses to transfect cells has recently been extended to show the utility of this procedure in vivo. Electrotransfer has been performed in vivo on several tissue types including skin, blood vessels, liver, tumor, muscle, cornea, brain and spleen. The most widely targeted tissue has been skeletal muscle. In addition to its potential use in gene therapy, in vivo DNA electrotransfer is also, because of its simplicity, a powerful laboratory tool to study in vivo gene expression and function in a given tissue. Many published studies have now shown that plasmid electrotransfer can lead to a long-lasting therapeutic effect in various pathologies, such as cancer, blood disease, or muscle ischemia. The future potential for this gene therapy approach will include delivery for both local action or distal effect by secretion of the transgenic proteins in the circulation.
Collapse
Affiliation(s)
- Daniel Scherman
- Laboratoire de Chimie Bioorganique, UMR 7001 ENSCP/CNRS/Gencell SA, CRVA-Aventis, Batiment Monod. 13 quai Jules Guesde, 94403 Vitry-sur-Seine cedex, France.
| | | | | |
Collapse
|
38
|
Heller LC, Coppola D. Electrically mediated delivery of vector plasmid DNA elicits an antitumor effect. Gene Ther 2002; 9:1321-5. [PMID: 12224015 DOI: 10.1038/sj.gt.3301802] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2001] [Accepted: 05/05/2002] [Indexed: 02/04/2023]
Abstract
In vivo electroporation is an efficient means of increasing plasmid DNA delivery to normal tissues, such as skin and muscle, as well as directly to tumors. In the experiments described here, plasmid DNA was delivered by in vivo electroporation to B16 mouse melanomas using two very different pulsing protocols. Reporter expression increased 21- or 42-fold, respectively with electroporation over injection alone. The growth of experimental melanomas with an approximate diameter of 4 mm on the day of treatment was monitored after electroporation delivery of reporter plasmid DNA. Remarkably, short-term complete regressions using one of these pulsing protocols occurred in up to 100% of mice. These regressions continued long term in up to 83% of animals. 70% of these mice were resistant to challenge with B16 melanoma cells. Histological analysis revealed large numbers of apoptotic cells 24 h after treatment. This antitumor effect did not require therapeutic cDNA expression or eukaryotic sequences.
Collapse
Affiliation(s)
- L C Heller
- Center for Molecular Delivery, c/o Department of Surgery MDC16, University of South Florida, 12901 Bruce B Downs Boulevard, Tampa, FL 33612, USA
| | | |
Collapse
|
39
|
Lefesvre P, Attema J, van Bekkum D. A comparison of efficacy and toxicity between electroporation and adenoviral gene transfer. BMC Mol Biol 2002; 3:12. [PMID: 12175426 PMCID: PMC122059 DOI: 10.1186/1471-2199-3-12] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2002] [Accepted: 08/13/2002] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Electroporation of skeletal muscle after injection of naked DNA was shown by others to increase transgene expression. Information regarding tissue damage caused by electroporation is conflicting. It is also not well known how plasmid electroporation compares with transfection by adenoviral vectors. To investigate these questions the most used protocol for muscle electroporation was used, i.e. 8 pulses of 200 V/cm and 20 ms at a frequency of 1 Hz. RESULTS Intra-muscular DNA transfer of pLuciferase was increased by 2 logs after electroporation, confirming data described by others. However, the blood levels of the encoded protein were still lower than those obtained after injection of first generation adenoviral vectors. Also, the electroporation procedure, on its own, caused severe muscle damage consisting of rhabdomyolysis and infiltration, whereas the adenoviral vectors caused only a slight infiltration. As damage of targeted tissue may be an advantage in the case of tumour transfection, we also compared the two transfection methods in tumour tissue. In case of poorly permissive tumours, adenoviral vectors cannot transfect more than 2% of the tumour tissue without inducing significant liver damage. In contrast, the electroporation seems to offer a wider therapeutic window since it does not cause any systemic toxicity and still induce's significant transfection. CONCLUSIONS Plasmid electroporation of the muscle induce severe local damage and is of no advantage over adenoviral vectors for obtaining high blood levels of a vector encoded protein. In contrast, electroporation of tumours might be safer than adenoviral gene transfer.
Collapse
Affiliation(s)
| | - Joline Attema
- Crucell BV, PO BOX 2048, 2301CA, Leiden, The Netherlands
| | | |
Collapse
|
40
|
Abstract
Adoptive cellular therapy remains a potentially powerful method of eradicating established tumors. T-cells have been particularly potent effector cells, as demonstrated in animal models and clinical studies, and it is apparent that the stimulation of certain subpopulations of T-cells that are reactive to tumor antigens can lead to more therapeutic T-cells. The use of gene transfer techniques has resulted in more effective and specific methods to generate these tumor-specific T-cells. Another area of tremendous interest is in the adoptive transfer of DCs manipulated to present tumor antigen to resting, naive T-cells. Gene transfer techniques may offer more optimal ways to generate therapeutic DCs. Adoptive immunotherapy may ultimately [figure: see text] have its greatest use in patients undergoing cellular rescue after ablative chemotherapy; the infusion of immunocompetent T-cells, genetically modified stem cells, or programmed DCs may offer the opportunity to direct a patient's immune response to eliminate residual microscopic disease.
Collapse
Affiliation(s)
- Alicia Terando
- Division of Surgical Oncology, University of Michigan Medical Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | | |
Collapse
|