1
|
Moaven O, W Mangieri C, A Stauffer J, Anastasiadis PZ, Borad MJ. Evolving Role of Oncolytic Virotherapy: Challenges and Prospects in Clinical Practice. JCO Precis Oncol 2021; 5:PO.20.00395. [PMID: 34250386 DOI: 10.1200/po.20.00395] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/04/2021] [Accepted: 01/27/2021] [Indexed: 12/23/2022] Open
Abstract
Selective oncotropism and cytolytic activity against tumors have made certain viruses subject to investigation as novel treatment modalities. However, monotherapy with oncolytic viruses (OVs) has shown limited success and modest clinical benefit. The capacity to genetically engineer OVs makes them a desirable platform to design complementary treatment modalities to overcome the existing treatment options' shortcomings. In recent years, our knowledge of interactions of the tumors with the immune system has expanded profoundly. There is a growing body of literature supporting immunomodulatory roles for OVs. The concept of bioengineering these platforms to induce the desired immune response and complement the current immunotherapeutic modalities to make immune-resistant tumors responsive to immunotherapy is under investigation in preclinical and early clinical trials. This review provides an overview of attempts to optimize oncolytic virotherapy as essential components of the multimodality anticancer therapeutic approach and discusses the challenges in translation to clinical practice.
Collapse
Affiliation(s)
- Omeed Moaven
- Section of Surgical Oncology, Department of Surgery, Mayo Clinic Florida, Jacksonville, FL
| | - Christopher W Mangieri
- Section of Surgical Oncology, Department of Surgery, Wake Forest University, Winston-Salem, NC
| | - John A Stauffer
- Section of Surgical Oncology, Department of Surgery, Mayo Clinic Florida, Jacksonville, FL
| | | | - Mitesh J Borad
- Division of Medical Oncology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ
| |
Collapse
|
2
|
Suzuki Y, Fujioka K, Ikeda K, Murayama Y, Manome Y. Temozolomide does not influence the transcription or activity of matrix metalloproteinases 9 and 2 in glioma cell lines. J Clin Neurosci 2017; 41:144-149. [PMID: 28408249 DOI: 10.1016/j.jocn.2017.03.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/13/2017] [Indexed: 10/19/2022]
Abstract
Glioblastoma multiforme (GBM) is a treatment-resistant malignancy with poor prognosis. Temozolomide (TMZ) is widely used as a first-line drug for GBM. Although this improves patient prognosis, it does not completely eradicate the tumour. Even after total surgical resection, GBM can exhibit uncontrollable invasiveness at the tumour margins owing to activation of matrix metalloproteinases (MMPs) such as MMP-2 and -9; these degrade collagen IV in the basement membrane, which normally prevents cancer invasion. TMZ induces DNA damage and activates transcription factors including c-jun, c-fos, nuclear factor-κβ, and early growth response protein-1, which have putative binding sites on the MMP-9 promoter. TMZ may therefore enhance tumour invasion by stimulating MMP-9 transcription and enzymatic activity. To test this hypothesis, we investigated MMP-2 and -9 mRNA transcription and activity in GBM cell lines treated with TMZ. Human A172 GBM cells were exposed to TMZ (25% and 50% inhibitory concentrations) for 24 or 48h; cell cycle distribution and mRNA levels of MMP-2 and -9 were evaluated using flow cytometry and semi-quantitative reverse transcription PCR, respectively. MMP-2 and -9 enzymatic activities were assessed using gelatin zymography in human A172 and U373 MG GBM cells exposed to TMZ under the same conditions. TMZ altered A172 cell cycle distribution, but not MMP-2 or -9 mRNA levels. TMZ did not affect MMP-2 or -9 enzymatic activities in A172 or U373 MG cells. These findings indicated that TMZ is therefore unlikely to promote GBM invasiveness.
Collapse
Affiliation(s)
- Yuta Suzuki
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, The Jikei University School of Medicine, Tokyo, Japan; Department of Neurosurgery, The Jikei University School of Medicine, Tokyo, Japan.
| | - Kouki Fujioka
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Keiichi Ikeda
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshinobu Manome
- Division of Molecular Cell Biology, Core Research Facilities for Basic Science, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
3
|
Tang Q, Lu M, Chen D, Liu P. Combination of PEI-Mn0.5Zn0.5Fe2O4 nanoparticles and pHsp 70-HSV-TK/GCV with magnet-induced heating for treatment of hepatoma. Int J Nanomedicine 2015; 10:7129-43. [PMID: 26604760 PMCID: PMC4655962 DOI: 10.2147/ijn.s92179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background To explore a new combination of thermal treatment and gene therapy for hepatoma, a heat-inducible herpes simplex virus thymidine kinase/ganciclovir (HSV-TK/GCV) gene therapy system was developed in which thermal energy generated by Mn0.5Zn0.5Fe2O4 nanoparticles (MZF-NPs) under an alternating magnetic field was used to activate gene expression. Methods First, a recombinant eukaryotic plasmid, pHsp 70-HSV-TK, was constructed as a target gene for therapy. This recombinant plasmid was used to transfect SMMC-7721 hepatoma cells and the gene expression was evaluated. Magnet-induced heating was then applied to cells to assess the antihepatoma effects of the polyethylenimine (PEI)-MZF-NPs/pHsp 70-HSV-TK/GCV complex, in vitro and in vivo. Results The results showed that cells were successfully transfected with pHsp 70-HSV-TK and that expression levels of HSV-TK remained stable. Both in vitro and in vivo results indicated that the combination of gene therapy and heat treatment resulted in better therapeutic effects than heating-alone group. The rates of apoptosis and necrosis in the combined treatment group were 49.0% and 7.21%, respectively. The rate of inhibition of cell proliferation in the combined treatment group was significantly higher (87.5%) than that in the heating-alone group (65.8%; P<0.01). The tumor volume and mass inhibition rates of the combined treatment group were 91.3% and 87.91%, respectively, and were significantly higher than the corresponding rates of the heating-alone group (70.41% and 57.14%; P<0.01). The expression levels of Stat3 and Bcl-xL messenger RNA and p-Stat3 and Bcl-xL protein in the combined treatment group were significantly lower than those in the other groups (P<0.01). The expression levels of Bax messenger RNA and protein in the recombinant plasmid group were significantly higher than those in the other groups (P<0.01). Conclusion It can therefore be concluded that the combined application of heat treatment and gene therapy has a synergistic and complementary effect and that PEI-MZF-NPs can simultaneously act both as a nonviral gene vector and a magnet-induced source of heat, thereby representing a viable approach for the treatment of cancer.
Collapse
Affiliation(s)
- Qiusha Tang
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| | - Mudan Lu
- Genetic Laboratory, Wuxi Hospital for Maternal and Child Health Care, the Affiliated Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Daozhen Chen
- Genetic Laboratory, Wuxi Hospital for Maternal and Child Health Care, the Affiliated Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Peidang Liu
- School of Medicine, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
4
|
Li L, Zhang CL, Kang L, Wang RF, Yan P, Zhao Q, Yin L, Guo FQ. Enhanced EJ Cell Killing of (125)I Radiation by Combining with Cytosine Deaminase Gene Therapy Regulated by Synthetic Radio-Responsive Promoter. Cancer Biother Radiopharm 2015; 30:342-8. [PMID: 26382009 PMCID: PMC4601673 DOI: 10.1089/cbr.2015.1862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aim: To investigate the enhancing effect of radionuclide therapy by the therapeutic gene placed under the control of radio-responsive promoter. Methods: The recombinant lentivirus E8-codA-GFP, including a synthetic radiation-sensitive promoter E8, cytosine deaminase (CD) gene, and green fluorescent protein gene, was constructed. The gene expression activated by 125I radiation was assessed by observation of green fluorescence. The ability of converting 5-fluorocytosine (5-FC) to 5-fluorourial (5-FU) by CD enzyme was assessed by high-performance liquid chromatography. The viability of the infected cells exposed to 125I in the presence of 5-FC was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and the infected cells exposed to 125I alone served as negative control and 5-FU as positive control. Results: The recombinant lentiviral vector was constructed successfully. On exposure of infected cells to 125I, green fluorescence can be observed and 5-FU can be detected. MTT assay showed that the survival rate for infected cells treated with 125I was lower compared with the 125I control group, but higher than the positive control group. Conclusion: The synthetic promoter E8 can induce the expression of downstream CD gene under 125I radiation, and the tumor killing effect of 125I can be enhanced by combining CD gene therapy with radiosensitive promoter.
Collapse
Affiliation(s)
- Ling Li
- 1 Department of Nuclear Medicine, Peking University First Hospital , Beijing, China
| | - Chun-li Zhang
- 1 Department of Nuclear Medicine, Peking University First Hospital , Beijing, China .,2 Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine , Wuxi, China
| | - Lei Kang
- 1 Department of Nuclear Medicine, Peking University First Hospital , Beijing, China
| | - Rong-Fu Wang
- 1 Department of Nuclear Medicine, Peking University First Hospital , Beijing, China
| | - Ping Yan
- 1 Department of Nuclear Medicine, Peking University First Hospital , Beijing, China
| | - Qian Zhao
- 1 Department of Nuclear Medicine, Peking University First Hospital , Beijing, China
| | - Lei Yin
- 1 Department of Nuclear Medicine, Peking University First Hospital , Beijing, China
| | - Feng-qin Guo
- 1 Department of Nuclear Medicine, Peking University First Hospital , Beijing, China
| |
Collapse
|
5
|
Abstract
In this article, advances in designing polymeric nanoparticles for targeted cancer gene therapy are reviewed. Characterization and evaluation of biomaterials, targeting ligands, and transcriptional elements are each discussed. Advances in biomaterials have driven improvements to nanoparticle stability and tissue targeting, conjugation of ligands to the surface of polymeric nanoparticles enable binding to specific cancer cells, and the design of transcriptional elements has enabled selective DNA expression specific to the cancer cells. Together, these features have improved the performance of polymeric nanoparticles as targeted non-viral gene delivery vectors to treat cancer. As polymeric nanoparticles can be designed to be biodegradable, non-toxic, and to have reduced immunogenicity and tumorigenicity compared to viral platforms, they have significant potential for clinical use. Results of polymeric gene therapy in clinical trials and future directions for the engineering of nanoparticle systems for targeted cancer gene therapy are also presented.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David R. Wilson
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camila G. Zamboni
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jordan J. Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Herman JM, Wild AT, Wang H, Tran PT, Chang KJ, Taylor GE, Donehower RC, Pawlik TM, Ziegler MA, Cai H, Savage DT, Canto MI, Klapman J, Reid T, Shah RJ, Hoffe SE, Rosemurgy A, Wolfgang CL, Laheru DA. Randomized phase III multi-institutional study of TNFerade biologic with fluorouracil and radiotherapy for locally advanced pancreatic cancer: final results. J Clin Oncol 2013; 31:886-894. [PMID: 23341531 PMCID: PMC4820756 DOI: 10.1200/jco.2012.44.7516] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE TNFerade biologic is a novel means of delivering tumor necrosis factor alpha to tumor cells by gene transfer. We herein report final results of the largest randomized phase III trial performed to date among patients with locally advanced pancreatic cancer (LAPC) and the first to test gene transfer against this malignancy. PATIENTS AND METHODS In all, 304 patients were randomly assigned 2:1 to standard of care plus TNFerade (SOC + TNFerade) versus standard of care alone (SOC). SOC consisted of 50.4 Gy in 28 fractions with concurrent fluorouracil (200 mg/m(2) per day continuous infusion). TNFerade was injected intratumorally before the first fraction of radiotherapy each week at a dose of 4 × 10(11) particle units by using either a percutaneous transabdominal or an endoscopic ultrasound approach. Four weeks after chemoradiotherapy, patients began gemcitabine (1,000 mg/m(2) intravenously) with or without erlotinib (100 to 150 mg per day orally) until progression or toxicity. RESULTS The analysis included 187 patients randomly assigned to SOC + TNFerade and 90 to SOC by using a modified intention-to-treat approach. Median follow-up was 9.1 months (range, 0.1 to 50.5 months). Median survival was 10.0 months for patients in both the SOC + TNFerade and SOC arms (hazard ratio [HR], 0.90; 95% CI, 0.66 to 1.22; P = .26). Median progression-free survival (PFS) was 6.8 months for SOC + TNFerade versus 7.0 months for SOC (HR, 0.96; 95% CI, 0.69 to 1.32; P = .51). Among patients treated on the SOC + TNFerade arm, multivariate analysis showed that TNFerade injection by an endoscopic ultrasound-guided transgastric/transduodenal approach rather than a percutaneous transabdominal approach was a risk factor for inferior PFS (HR, 2.08; 95% CI, 1.06 to 4.06; P = .032). The patients in the SOC + TNFerade arm experienced more grade 1 to 2 fever and chills than those in the SOC arm (P < .001) but both arms had similar rates of grade 3 to 4 toxicities (all P > .05). CONCLUSION SOC + TNFerade is safe but not effective for prolonging survival in patients with LAPC.
Collapse
Affiliation(s)
- Joseph M Herman
- Department of Radiation Oncology & Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, 401 N. Broadway, Weinberg Suite 1440, Baltimore, MD 21231, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Touchefeu Y, Vassaux G, Harrington KJ. Oncolytic viruses in radiation oncology. Radiother Oncol 2011; 99:262-70. [PMID: 21704402 DOI: 10.1016/j.radonc.2011.05.078] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 05/27/2011] [Accepted: 05/28/2011] [Indexed: 10/18/2022]
Abstract
Oncolytic viruses are investigational cancer treatments. They are currently being assessed as single agents or in combination with standard therapies such as external beam radiotherapy - a DNA damaging agent that is a standard of care for many tumour types. Preclinical data indicate that combinations of oncolytic viruses and radiation therapy are promising, showing additional or synergistic antitumour effects in in vitro and in vivo studies. This interaction has the potential to be multifaceted: viruses may act as radiosensitizing agents, but radiation may also enhance viral oncolysis by increasing viral uptake, replication, gene expression and cell death (apoptosis, autophagy or necrosis) in irradiated cells. Phase I and II clinical trials investigating combinations of viruses and radiation therapy have been completed, paving the way for ongoing phase III studies. The aim of this review is to focus on the therapeutic potential of these combinations and to highlight their mechanistic bases, with particular emphasis on the role of the DNA damage response.
Collapse
Affiliation(s)
- Yann Touchefeu
- The Institute of Cancer Research, Section of Cell and Molecular Biology, London, UK
| | | | | |
Collapse
|
8
|
Guo R, Tian L, Han B, Xu H, Zhang M, Li B. Feasibility of a novel positive feedback effect of 131I-promoted Bac-Egr1-hNIS expression in malignant glioma via baculovirus. Nucl Med Biol 2011; 38:599-604. [DOI: 10.1016/j.nucmedbio.2010.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2010] [Revised: 11/14/2010] [Accepted: 11/16/2010] [Indexed: 11/29/2022]
|
9
|
Yang W, Zhao JG, Li XY, Gong SL, Cao JP. Anti-tumor effect of 125I-UdR in combination with Egr-1 promoter-based IFNγ gene therapy in vivo. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2010; 49:705-714. [PMID: 20602107 DOI: 10.1007/s00411-010-0312-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Accepted: 06/20/2010] [Indexed: 05/29/2023]
Abstract
Although (125)I-UdR treatment of malignant tumors in animal models and patients has achieved a certain effect, the short half-life of (125)I-UdR in vivo and its cellular uptake only in S phase of the cell cycle are limiting factors with regard to tumor eradication, and therefore its combination with other applications is a promising strategy in cancer therapy. In this study, we show that (125)I-UdR radionuclide therapy in combination with Egr-1 promoter-based IFNγ gene therapy is more effective than (125)I-UdR therapy alone in suppressing tumor growth and extending survival duration in mice bearing H22 hepatomas. Combined therapy could significantly inhibit cell proliferation and tumor angiogenesis, induce apoptosis and enhance cytotoxic activities of splenic CTL of the mice. Our results suggest that (125)I-UdR in combination with Egr-1 promoter-based IFNγ gene therapy may provide novel approaches for cancer treatment.
Collapse
Affiliation(s)
- Wei Yang
- Department of Radiobiology, School of Radiological Medicine and Public Health, Soochow University, Suzhou, China.
| | | | | | | | | |
Collapse
|
10
|
Murugesan SR, King CR, Osborn R, Fairweather WR, O'Reilly EM, Thornton MO, Wei LL. Combination of human tumor necrosis factor-alpha (hTNF-alpha) gene delivery with gemcitabine is effective in models of pancreatic cancer. Cancer Gene Ther 2009; 16:841-7. [PMID: 19444305 DOI: 10.1038/cgt.2009.32] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Pancreatic adenocarcinoma is an aggressive and highly lethal malignancy. Currently, gemcitabine is commonly used in patients with pancreatic cancer. However, the life expectancy of pancreatic cancer patients remains poor. We explored the possibility of increased anti-tumor activity by combining human tumor necrosis factor-alpha (hTNF-alpha) with current front-line therapy. Human TNF-alpha displays potent anti-tumor activity, but its use is limited by the toxicity of systemic administration. We developed a gene delivery approach using intratumoral injections of an adenoviral vector expressing hTNF-alpha, AdEgr.TNF.11D (TNFerade), to increase local concentrations of hTNF-alpha within the tumor, thereby maximizing local anti-tumor activity and yet minimizing the systemic toxicities. An ongoing phase III clinical trial is testing the efficacy of AdEgr.TNF.11D-injected intratumorally and combining with chemotherapy in locally advanced pancreatic cancer. In this study, we show that treatment with AdEgr.TNF.11D and gemcitabine results in a high level of hTNF-alpha expression in human pancreatic cancer cell lines. The combined treatment was well tolerated, highly active and produced marked delays in the growth of human pancreatic xenograft tumors relative to either agent alone. Our results strongly suggest that combination of AdEgr.TNF.11D and gemcitabine may be a potentially useful therapeutic approach for the improved treatment of pancreatic cancer.
Collapse
Affiliation(s)
- S R Murugesan
- Department of Research, GenVec Inc, Gaithersburg, MD 20878, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Systemic therapy for cervical cancer with potentially regulatable oncolytic adenoviruses. PLoS One 2008; 3:e2917. [PMID: 18698374 PMCID: PMC2500220 DOI: 10.1371/journal.pone.0002917] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Accepted: 07/07/2008] [Indexed: 12/20/2022] Open
Abstract
Clinical trials have confirmed the safety of selectively oncolytic adenoviruses for treatment of advanced cancers. However, increasingly effective viruses could result in more toxicity and therefore it would be useful if replication could be abrogated if necessary. We analyzed viruses containing the cyclooxygenase-2 (Cox-2) or vascular endothelial growth factor (VEGF) promoter for controlling replication. Anti-inflammatory agents can lower Cox-2 protein levels and therefore we hypothesized that also the promoter might be affected. As Cox-2 modulates expression of VEGF, also the VEGF promoter might be controllable. First, we evaluated the effect of anti-inflammatory agents on promoter activity or adenovirus infectivity in vitro. Further, we analyzed the oncolytic potency of the viruses in vitro and in vivo with and without the reagents. Moreover, the effect of on virus replication was analyzed. We found that RGD-4C or Ad5/3 modified fibers improved the oncolytic potency of the viruses in vitro and in vivo. We found that both promoters could be downregulated with dexamethasone, sodium salicylate, or salicylic acid. Oncolytic efficacy correlated with the promoter activity and in vitro virus production could be abrogated with the substances. In vivo, we saw good therapeutic efficacy of the viruses in a model of intravenous therapy of metastatic cervical cancer, but the inhibitory effect of dexamethasone was not strong enough to provide significant differences in a complex in vivo environment. Our results suggest that anti-inflammatory drugs may affect the replication of adenovirus, which might be relevant in case of replication associated side effects.
Collapse
|
12
|
Tang QS, Zhang DS, Cong XM, Wan ML, Jin LQ. Using thermal energy produced by irradiation of Mn–Zn ferrite magnetic nanoparticles (MZF-NPs) for heat-inducible gene expression. Biomaterials 2008; 29:2673-9. [DOI: 10.1016/j.biomaterials.2008.01.038] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Accepted: 01/25/2008] [Indexed: 11/30/2022]
|
13
|
|
14
|
Ma HB, Wang XJ, Di ZL, Xia H, Li Z, Liu J, Ma J, Kang HF, Wu CM, Bai MH. Construction of targeted plasmid vector pcDNA3.1-Egr.1p-p16 and its expression in pancreatic cancer JF305 cells induced by radiation in vitro. World J Gastroenterol 2007; 13:4214-8. [PMID: 17696250 PMCID: PMC4250620 DOI: 10.3748/wjg.v13.i31.4214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct pcDNA3.1-Egr.1p-p16 recombinant plasmid and investigate the expression of p16 in pancreatic cancer JF305 cells induced by radiation and the feasibility of gene radiotherapy for pancreatic carcinoma.
METHODS: Human p16 cDNA was ligated to the downstream of Egr-1 promotor to construct pcDNA3.1-Egr.1p-p16 plasmid by restriction enzyme digested. The recombined plasmids were transfected into pancreatic cancer JF305 cells with lipofectamine. p16 mRNA level was detected by RT-PCR. The expression of p16 after different doses of X-ray radiation was detected by Western blot technique. Cell survival was assessed by clonogenic assays and cell viability was analysed by trypen blue exclusion. Flow cytometry was performed to study the apoptosis of JF305 cells.
RESULTS: Restriction enzyme digestion showed the correctly constructed pcDNA3.1-Egr.1p-p16. The p16 expression in cells transfected with pcDNA3.1-Egr.1p-p16 induced by different doses of radiation was higher than that in the control group (P < 0.05). Eight hours after 2 Gy X-ray radiation, the expression reached its peak (87.00 ng/L), and was significantly higher than that in the control group (P < 0.0.5). Clonogenic analysis and trypan blue extraction test showed that the pcDNA3.1-Egr.1p-p16 transfer enhanced radiation-induced cell killing in p16-null JF305 cell lines. The induction of apoptosis was lower in combined transfection and irradiation group than that in irradiation alone.
CONCLUSION: X-ray can induce the recombinant plasmid pcDNA3.1-Egr.1p-p16 expression in JF305 cells. The detection of dose and time provides an experimental basis for in vivo study in future.
Collapse
Affiliation(s)
- Hong-Bing Ma
- Department of Oncology, Second Hospital, Medical college of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Tsurushima H, Yuan X, Dillehay LE, Leong KW. Radioresponsive tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene therapy for malignant brain tumors. Cancer Gene Ther 2007; 14:706-16. [PMID: 17541421 DOI: 10.1038/sj.cgt.7701065] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Patients with malignant gliomas have a very poor prognosis. To explore a novel and more effective approach for the treatment of malignant gliomas, a strategy that combined tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene therapy and radiation treatment (RT) was designed in this study. Plasmid pE4-GFP was constructed by including the radioinducible early growth response gene 1 (Egr-1) promoter, and it yielded the best response with fractionated RT. Plasmid pE4-TRAIL was constructed by including the Egr-1 promoter and evaluated using U251 and U87 glioma cells. In the assay of apoptosis and killing activities, pE4-TRAIL exhibited radioresponse. pE4-TRAIL combined with RT is capable of inducing cell death synergistically. The expression of TRAIL death receptors was evaluated; which may be influenced by RT. Glioma cells with wild-type p53 showed upregulated expression of death receptors, and more synergistic effects on killing activities are expected. pE4-TRAIL was transfected into the subcutaneous U251 glioma cells in nude mice by the in vivo electroporation method. In the mice treated with pE4-TRAIL and RT, apoptotic cells were detected in pathological sections, and a significant difference of tumor volumes was observed when compared with the other groups (P<0.001). Our results indicate that radioresponsive gene therapy may have great potential as a novel therapy because this therapeutic system can be spatially or temporally controlled by exogenous RT and provides specificity and safety.
Collapse
Affiliation(s)
- H Tsurushima
- Department of Biomedical Engineering, Medical School, Johns Hopkins University, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
16
|
Tsurushima H, Yuan X, Dillehay LE, Leong KW. Radio-responsive gene therapy for malignant glioma cells without the radiosensitive promoter: Caspase-3 gene therapy combined with radiation. Cancer Lett 2007; 246:318-23. [PMID: 16644107 DOI: 10.1016/j.canlet.2006.03.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Accepted: 03/13/2006] [Indexed: 12/13/2022]
Abstract
Caspase-3 plays a critical role as an executioner of apoptosis. The aim of this study is to evaluate the potential of the combination of caspase-3 gene therapy and radiation treatment. We prepared a plasmid (pCI-CSP3) that contained the human caspase-3 gene and the cytomegalovirus promoter. We introduced this plasmid into U251 and U87 human glioma cells and subjected the cells to radiation treatment. The degree of cell death and apoptosis were evaluated. None of the cell lines underwent apoptosis by the overexpression of caspase-3 alone, but the degree of cell death and apoptosis were markedly enhanced by the addition of radiation treatment. Next, we prepared another plasmid (EGR-CSP3) that contained the caspase-3 gene and a radiation-sensitive promoter. Each treatment system using either pCI-CSP3 or EGR-CSP3 showed radio response. The treatment system using pCI-CSP3 more effectively induced apoptosis than that using EGR-CSP3. Caspase-3 gene therapy in combination with radiation treatment has the potential to serve as a radio-responsive gene therapy without any radiation-sensitive promoter.
Collapse
Affiliation(s)
- Hideo Tsurushima
- Department of Biomedical Engineering, Medical School, Johns Hopkins University, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
17
|
Lumniczky K, Sáfrány G. Cancer gene therapy: Combination with radiation therapy and the role of bystander cell killing in the anti-tumor effect. Pathol Oncol Res 2006; 12:118-24. [PMID: 16799716 DOI: 10.1007/bf02893457] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2005] [Accepted: 04/20/2006] [Indexed: 11/28/2022]
Abstract
Current anti-cancer modalities such as surgery, chemo- and radiation therapies have only limited success in cancer treatment. Gene therapy is a promising new tool to improve outcomes. In this review, first we summarize the various strategies to kill tumor cells, and then focus on the bystander effect of gene therapy. A variety of strategies, such as gene-directed enzyme pro-drug therapy, activation of an anti-tumor immune attack, application of replication-competent and oncolytic viral vectors, tumor-specific as well as radiation- and hypoxiainduced gene expression, might be applied to target tumor cells. We put special emphasis on the combination of these approaches with local tumor irradiation. Using the available vector systems, only a small portion of cancer cells contains the therapeutic genes under clinical situations. However, cells directly targeted by gene therapy will transfer death signals to neighboring cancer cells. This bystander cell killing improves the efficiency of cancer gene therapy. Death signals are delivered by cell-to-cell communication through gap junction intercellular contacts, release of toxic metabolites into the neighborhood or to larger distances, phagocytosis of apoptotic bodies, and the activation of the immune system. Bystander cell killing can be enhanced by the introduction of gap junction proteins into cells, by further activating the immune system with immune-stimulatory molecules, or by introducing genes that help the transfer of cytotoxic genes and/or metabolites into bystander cells. In conclusion, although bystander cell killing can improve therapeutic effects, there should be additional developments in cancer gene therapy for a more efficient clinical application.
Collapse
Affiliation(s)
- Katalin Lumniczky
- Department of Molecular and Tumor Radiobiology, National Research Institute for Radiobiology and Radiohygiene, Budapest, H-1221, Hungary
| | | |
Collapse
|
18
|
Glinka EM, Edelweiss EF, Deyev SM. Eukaryotic expression vectors and immunoconjugates for cancer therapy. BIOCHEMISTRY (MOSCOW) 2006; 71:597-606. [PMID: 16827650 DOI: 10.1134/s0006297906060022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This review considers ways to address specificity to therapeutic targeted anticancer agents. These include transcriptional activation of tissue- and tumor-specific promoters in eukaryotic expression vectors and use of antitumor-directed immunoconjugates. The review deals with analysis of strategies used for selection of targeted promoters and examples of antibody fusion proteins exhibiting antitumor activity. A new direction in antitumor treatment pooling together methods of gene therapy and antibody therapy has appeared. This direction is based on the development of vectors encoding secreted forms of immunoconjugates. After vector introduction into a cell, the latter is capable of synthesizing and secreting antibody fusion protein composed of a therapeutic anticancer agent and antibody specifically targeted to cancer cells.
Collapse
Affiliation(s)
- E M Glinka
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
| | | | | |
Collapse
|
19
|
Van Houdt WJ, Haviv YS, Lu B, Wang M, Rivera AA, Ulasov IV, Lamfers MLM, Rein D, Lesniak MS, Siegal GP, Dirven CMF, Curiel DT, Zhu ZB. The human survivin promoter: a novel transcriptional targeting strategy for treatment of glioma. J Neurosurg 2006; 104:583-92. [PMID: 16619663 DOI: 10.3171/jns.2006.104.4.583] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Malignant brain tumors have been proved to be resistant to standard treatments and therefore require new therapeutic strategies. Survivin, a recently described member of the inhibitor of apoptosis protein family, is overexpressed in several human brain tumors, primarily gliomas, but is downregulated in normal tissues. The authors hypothesized that the expression of tumor-specific survivin could be exploited for treatment of gliomas by targeting the tumors with gene therapy vectors.
Methods
Following confirmation of survivin expression in glioma cell lines, an adenoviral vector containing the survivin promoter and the reporter gene luciferase was tested in established and primary glioma cells, normal astrocytic cells, and normal human brain tissues. High levels of reporter gene expression were observed in established tumor and primary tumor cell lines and low levels of expression in astrocytes and normal human brain tissue. To test oncolytic potency, the authors constructed survivin promoter–based conditionally replicative adenoviruses (CRAds), composed of survivin promoter–regulated E1 gene expression and an RGD-4C capsid modification. These CRAds could efficiently replicate within and kill a variety of established glioma tumor cells, but were inactive in a normal human liver organ culture. Finally, survivin promoter–based CRAds significantly inhibited the growth of glioma xenografts in vivo.
Conclusions
Together these data indicate that the survivin promoter is a promising tumor-specific promoter for transcriptional targeting of adenovirus-based vectors and CRAds for malignant gliomas. The strategy of using survivin–CRAds may thus translate into an experimental therapeutic approach that can be used in human clinical trials.
Collapse
Affiliation(s)
- Winan J Van Houdt
- Department of Neurosurgery, VU Universiteit Medische Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Horsman MR, Bohm L, Margison GP, Milas L, Rosier JF, Safrany G, Selzer E, Verheij M, Hendry JH. Tumor radiosensitizers--current status of development of various approaches: report of an International Atomic Energy Agency meeting. Int J Radiat Oncol Biol Phys 2006; 64:551-61. [PMID: 16414371 DOI: 10.1016/j.ijrobp.2005.09.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Revised: 08/30/2005] [Accepted: 09/29/2005] [Indexed: 01/02/2023]
Abstract
PURPOSE The International Atomic Energy Agency (IAEA) held a Technical Meeting of Consultants to (1) discuss a selection of relatively new agents, not those well-established in clinical practice, that operated through a variety of mechanisms to sensitize tumors to radiation and (2) to compare and contrast their tumor efficacy, normal tissue toxicity, and status of development regarding clinical application. The aim was to advise the IAEA as to which developing agent or class of agents would be worth promoting further, by supporting additional laboratory research or clinical trials, with the eventual goal of improving cancer control rates using radiotherapy, in developing countries in particular. RESULTS The agents under discussion included a wide, but not complete, range of different types of drugs, and antibodies that interfered with molecules in cell signaling pathways. These were contrasted with new molecular antisense and gene therapy strategies. All the drugs discussed have previously been shown to act as tumor cell radiosensitizers or to kill hypoxic cells present in tumors. CONCLUSION Specific recommendations were made for more preclinical studies with certain of the agents and for clinical trials that would be suitable for industrialized countries, as well as trials that were considered more appropriate for developing countries.
Collapse
Affiliation(s)
- Michael R Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Mairs RJ, Fullerton NE, Cosimo E, Boyd M. Gene manipulation to enhance MIBG-targeted radionuclide therapy. Nucl Med Biol 2006; 32:749-53. [PMID: 16243651 DOI: 10.1016/j.nucmedbio.2005.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2005] [Revised: 03/22/2005] [Accepted: 03/24/2005] [Indexed: 11/18/2022]
Abstract
The goal of targeted radionuclide therapy is the deposition in malignant cells of sterilizing doses of radiation without damaging normal tissue. The radiopharmaceutical [(131)I]meta-iodobenzylguanidine ([(131)I]MIBG) is an effective single agent for the treatment of neuroblastoma. However, uptake of the drug in malignant sites is insufficient to cure disease. A growing body of experimental evidence indicates exciting possibilities for the integration of gene transfer with [(131)I]MIBG-targeted radiotherapy.
Collapse
Affiliation(s)
- Robert J Mairs
- Targeted Therapy Group, Centre for Oncology and Applied Pharmacology, Cancer Research UK Beatson Laboratories, University of Glasgow.
| | | | | | | |
Collapse
|
22
|
Glinka EM, Edelweiss EF, Sapozhnikov AM, Deyev SM. A new vector for controllable expression of an anti-HER2/neu mini-antibody-barnase fusion protein in HEK 293T cells. Gene 2006; 366:97-103. [PMID: 16300908 DOI: 10.1016/j.gene.2005.06.042] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2005] [Accepted: 06/27/2005] [Indexed: 11/18/2022]
Abstract
Tumor-targeted vectors with controllable expression of therapeutic genes and specific antitumor antibodies are promising tools for the reduction of malignant tumors. Here we describe a new plasmid for the eukaryotic expression of an anti-HER2/neu mini-antibody-barnase fusion protein (4D5 scFv-barnase-His(5)) with an NH(2)-terminal leader peptide. The 4D5 scFv-barnase-His(5) gene was placed downstream of the tetracycline responsive-element minimal promoter in the vector using the Tet-Off gene-expression system. The Bacillus amyloliquefaciens ribonuclease barnase is toxic for the host cells. To overcome this problem, barstar gene under its own minimal cytomegalovirus promoter was used in designed vector. Barstar inhibits the background level of barnase in the cells in the presence of tetracycline in culture medium. The HEK 293T cells were transfected with the designed vector, and the 4D5 scFv-barnase-His(5) fusion protein was identified by anti-barnase antibodies in cell culture medium and after purification from cell lysates using metal-affinity chromatography. The overexpression of the anti-HER2/neu mini-antibody-barnase fusion protein decreased the intensity of fluorescence of HEK 293T cells co-transfected with the generated plasmid and a plasmid containing the gene of enhanced green fluorescent protein (pEGFP-N1), in comparison with the intensity of fluorescence of HEK 293T cells transfected with pEGFP-N1, in the absence of tetracycline in the medium. The effect of the 4D5 scFv-barnase-His(5) on EGFP fluorescence indicates that the introduced barnase functions as a ribonuclease inside the cells. The anti-HER2/neu mini-antibody could be used to deliver barnase to HER2/neu-positive cells and provide its penetration into the target cells, as HER2/neu is a ligand-internalizing receptor. This expression vector has potential applications to both gene and antibody therapies of cancer.
Collapse
Affiliation(s)
- Elena M Glinka
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian, Academy of Sciences, Miklukho-Maklaya, 16/10, Moscow, 117997, Russia.
| | | | | | | |
Collapse
|
23
|
Anton M, Gomaa IEO, von Lukowicz T, Molls M, Gansbacher B, Würschmidt F. Optimization of radiation controlled gene expression by adenoviral vectors in vitro. Cancer Gene Ther 2005; 12:640-6. [PMID: 15803145 DOI: 10.1038/sj.cgt.7700829] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The radiation-inducible EGR-1-promoter has been used in different gene therapy approaches in order to enhance and locally restrict therapeutic efficacy. The aim of this study was to reduce nonspecific gene expression in the absence of irradiation (IR) in an adenoviral vector. Rat rhabdomyosarcoma R1H tumor cells were infected with adenoviral vectors expressing either EGFP or HSV-TK under control of the murine EGR-1 promoter/enhancer. Cells were irradiated at 0-6 Gy. Gene expression was determined by FACS-analysis (EGFP), or crystal violet staining (HSV-TK). The bovine growth hormone polyadenylation signal (BGH pA) was used as insulating sequence and was introduced upstream or upstream and downstream of the expression cassette. Infected R1H cells displayed IR dose-dependent EGFP expression. Cells treated with IR, AdEGR.TK and ganciclovir displayed a survival of 17.3% (6 Gy). However, significant gene expression was observed in the absence of IR with EGR.TK and EGR.EGFP constructs. Introduction of BGHpA upstream or upstream and downstream of expression cassette resulted in decreased nonspecific cytotoxicity by a factor of 1.6-2.3 with minor influence on the induced level of cytotoxicity. Introduction of insulating sequences in adenoviral vectors might allow tighter temporospatial control of gene expression by the radiation-inducible EGR-1 promoter.
Collapse
Affiliation(s)
- Martina Anton
- Institut für Experimentelle Onkologie & Therapieforschung, München, Germany.
| | | | | | | | | | | |
Collapse
|
24
|
Rubinchik S, Woraratanadharm J, Yu H, Dong JY. New complex Ad vectors incorporating both rtTA and tTS deliver tightly regulated transgene expression both in vitro and in vivo. Gene Ther 2005; 12:504-11. [PMID: 15660114 DOI: 10.1038/sj.gt.3302437] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Regulation of transgene expression is a major goal of gene therapy research. Previously, we have developed a complex adenovirus (Ad) vector with tetracycline-regulated expression of a Fas ligand (FasL)-green fluorescent protein (GFP) fusion protein. This vector delivered high levels of activity that was regulated by doxycycline. However, this regulation was limited by the low but significant background activity of the TRE promoter. Recently, the Tet-regulated transcriptional silencer, tTS, was reported to suppress efficiently basal TRE activity without affecting induced expression levels. Here, we report development of Ad vectors that incorporate tTS in combination with that of reverse transactivator (rtTA) coupled with TRE promoter driving transgene expression. Incorporation of tTS improved control of transgene expression in vitro, so that an induction range of over three orders of magnitude was achieved in some cell lines. Effective regulation of transgene expression was also seen in a mouse model in vivo, following systemic vector delivery. In the case of FasL-GFP expression, significant improvement in the control of apoptotic activity both in vitro and in a mouse hepatotoxicity model was demonstrated when using rtTA-tTS vectors. In conclusion, a highly effective transgene regulation system, deliverable by a single adenoviral vector, is now available.
Collapse
Affiliation(s)
- S Rubinchik
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29403, USA
| | | | | | | |
Collapse
|
25
|
Manome Y, Nakayama N, Nakayama K, Furuhata H. Insonation facilitates plasmid DNA transfection into the central nervous system and microbubbles enhance the effect. ULTRASOUND IN MEDICINE & BIOLOGY 2005; 31:693-702. [PMID: 15866419 DOI: 10.1016/j.ultrasmedbio.2005.01.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2004] [Revised: 01/19/2005] [Accepted: 01/27/2005] [Indexed: 05/02/2023]
Abstract
Many of the diseases which affect the central nervous system are intractable to conventional therapies and therefore require alternative treatments such as gene therapy. Therapy requires safety, since the central nervous system is a critical organ. Choice of nonviral vectors such as naked plasmid DNA may have merit. However, transfection efficiencies of these vectors are low. We have investigated the use of 210.4 kHz ultrasound and found that 5.0 W/cm(2) of insonation for 5 s most effectively transfected a plasmid DNA into culture slices of mouse brain (147.68-fold increase compared with 0 W/cm(2) of insonation for 5 s). The effect was reinforced by combination with echo contrast agent, Levovist. One hundred fifty mg/mL of Levovist significantly increased gene transfection by ultrasound (5.23-fold when insonated at 5.0 W/cm(2) for 5 s). When DNA was intracranially injected, Levovist also enhanced gene transfection in newborn mice (4.49-fold increase when insonated at 5.0 W/cm(2) for 5 s). Since ultrasound successfully transfected naked plasmid DNA into the neural tissue and Levovist enhanced the effect, this approach may have a significant role in gene transfer to the central nervous system.
Collapse
Affiliation(s)
- Yoshinobu Manome
- Department of Molecular Cell Biology, Institute of DNA Medicine, Research Center for Medical Science, Jikei University School of medicine, 3-25-8 Nishishinbashi, Minato-ku, Tokyo, Japan 105-8461.
| | | | | | | |
Collapse
|
26
|
|
27
|
|
28
|
Xu ZL, Mizuguchi H, Mayumi T, Hayakawa T. Regulated gene expression from adenovirus vectors: a systematic comparison of various inducible systems. Gene 2003; 309:145-51. [PMID: 12758130 DOI: 10.1016/s0378-1119(03)00506-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Positively and tightly regulated gene expression is essential for gene function and gene therapy research. The currently-used inducible gene expression systems include tetracycline (Tet-on and T-REx), ecdysone, antiprogestin and dimerizer-based systems. Adenovirus (Ad) vectors play an important role in gene function and gene therapy research for their various advantages over other vector systems. Previously, we reported the inferiority of the Tet-on system as an inducible gene expression system in the context of Ad vectors in comparison with the Tet-off system. In this study, to identify an optimal system for regulated gene expression from Ad vectors, we made a rigorous direct comparison of these five inducible gene expression systems in three cell lines using the luciferase reporter gene. The highest sensitivity to the respective inducer was that of the dimerizer system, followed by the antiprogestin system. The lowest basal expression and the highest induction factor were both characteristic of the dimerizer system. Furthermore, the dimerizer and T-REx systems exhibited much higher induced expression levels than the other three systems. The elucidation of the characteristic features of each system should provide important information for widespread and feasible application of these systems. Overall, these results suggest the most appropriate inducible gene expression system in the context of Ad vectors to be the dimerizer system.
Collapse
Affiliation(s)
- Zhi-Li Xu
- Division of Cellular and Gene Therapy Products, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | | | | | | |
Collapse
|
29
|
Abstract
Cancer gene therapy has been one of the most exciting areas of therapeutic research in the past decade. In this review, we discuss strategies to restrict transcription of transgenes to tumour cells. A range of promoters which are tissue-specific, tumour-specific, or inducible by exogenous agents are presented. Transcriptional targeting should prevent normal tissue toxicities associated with other cancer treatments, such as radiation and chemotherapy. In addition, the specificity of these strategies should provide improved targeting of metastatic tumours following systemic gene delivery. Rapid progress in the ability to specifically control transgenes will allow systemic gene delivery for cancer therapy to become a real possibility in the near future.
Collapse
Affiliation(s)
- Tracy Robson
- School of Biomedical Sciences, University of Ulster, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| | - David G. Hirst
- School of Biomedical Sciences, University of Ulster, Newtownabbey, Co. Antrim, BT37 0QB, Northern Ireland, UK
| |
Collapse
|
30
|
Guilhon E, Voisin P, de Zwart JA, Quesson B, Salomir R, Maurange C, Bouchaud V, Smirnov P, de Verneuil H, Vekris A, Canioni P, Moonen CTW. Spatial and temporal control of transgene expression in vivo using a heat-sensitive promoter and MRI-guided focused ultrasound. J Gene Med 2003; 5:333-42. [PMID: 12692867 DOI: 10.1002/jgm.345] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Among the techniques used to induce and control gene expression, a non-invasive, physical approach based on local heat in combination with a heat-sensitive promoter represents a promising alternative but requires accurate temperature control in vivo. MRI-guided focused ultrasound (MRI-FUS) with real-time feedback control allows automatic execution of a predefined temperature-time trajectory. The purpose of this study was to demonstrate temporal and spatial control of transgene expression based on a well-defined local hyperthermia generated by MRI-FUS. METHODS Expression of the green fluorescent protein (GFP) marker gene was used. Two cell lines were derived from C6 glioma cells. The GFP expression of the first one is under the control of the CMV promoter, whereas it is under the control of the HSP70 promoter in the second one and thus inducible by heat. Subcutaneous tumours were generated by injection in immuno-deficient mice and rats. Tumours were subjected to temperatures varying from 42 to 50 degrees C for 3 to 25 min controlled by MRI-FUS and analyzed 24 h after the heat-shock. Endogenous HSP70 expression and C6 cell distribution were also analyzed. RESULTS The results demonstrate strong expression at 50 degrees C applied during a short time period (3 min) without affecting cell viability. Induced expression was also clearly shown for temperature in the range 44-48 degrees C but not at 42 degrees C. CONCLUSIONS Heating with MRI-FUS allows a tight and non-invasive control of transgene expression in a tumour.
Collapse
Affiliation(s)
- E Guilhon
- Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS/Victor Segalen Université Victor Segalen Bordeaux 2, Bordeaux, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Du N, Feng K, Luo C, Li L, Bai C, Pei X. Radioprotective effect of FLT3 ligand expression regulated by Egr-1 regulated element on radiation injury of SCID mice. Exp Hematol 2003; 31:191-6. [PMID: 12644015 DOI: 10.1016/s0301-472x(02)01082-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Hematopoietic factors have an important effect on the regulation of hematopoiesis by stimulating the proliferation of hematopoietic progenitor cells. Although the cytokines that stimulate hematopoiesis have also often proved to exert radioprotective effects, no definitive correlation has been found between the expression of these cytokines regulated by radio-inducible genes and their radioprotective effects. In the current experiments, we evaluated the radioprotective effects of the hematopoietic growth factors regulated by a radio-inducible promoter on radiation injury. METHODS The human Flt3 (FL) cDNA and enhanced green fluorescent protein (EGFP) cDNA were linked together with the internal ribosome entry site (IRES) and then inserted into the eukaryotic expression vector pCI-neo with the Egr-1 promoter (Egr-GF), and the vector was transduced into bone marrow stromal cell lines HFCL (HFCL/EGF). The level of green fluorescence in HFCL/EGF cells was detected after radiation with flow cytometry. The expression of FL in irradiated HFCL/EGF cells was confirmed with Northern blot and ELISA. The HFCL/EGF and CD34(+) cells from human umbilical cord blood were sequentially transplanted intravenously into sublethally irradiated severe combined immunodeficient (SCID) mice. The numbers of peripheral white blood cells transplanted into recipient mice were detected. RESULTS The activity of EGFP in transfected cells was significantly increased after exposure to gamma radiation at 2.0, 2.5, and 5.0 Gy as compared with nontransfected cells. The expression of FL in HFCL/EGF was significantly higher than that of the control groups (HFCL, HFCL/pCI-neo, and HFCL/FG). The level of secreted FL in serum-free supernatants of HFCL/EGF on human CD34(+) cells was higher than that of control groups. In contrast with three control groups (HFCL, HFCL/pCI-neo, and HFCL/GF), HFCL/EGF resulted in a proportional increase in the number of white blood cells at an early stage after radiation. CONCLUSIONS We show that radiation enhances the ability of expression of FL in HFCL/EGF to stimulate the proliferation of hematopoietic progenitor cells. These results suggest in vivo use of gene therapy of FL regulated by the Egr-1 promoter protects hematopoiesis from irradiation-induced damage.
Collapse
Affiliation(s)
- Nan Du
- Department of Stem Cell Biology, Beijing Institute of Transfusion Medicine, Beijing, China
| | | | | | | | | | | |
Collapse
|
32
|
Weichselbaum RR, Kufe DW, Hellman S, Rasmussen HS, King CR, Fischer PH, Mauceri HJ. Radiation-induced tumour necrosis factor-alpha expression: clinical application of transcriptional and physical targeting of gene therapy. Lancet Oncol 2002; 3:665-71. [PMID: 12424068 DOI: 10.1016/s1470-2045(02)00900-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Promising data are emerging on a new anticancer agent, Ad.EGR-TNF, an adenoviral vector, which contains radio-inducible DNA sequences from the early growth response (EGR1) gene promoter and cDNA for the gene encoding human tumour necrosis factor-alpha. Ad.EGR-TNF combines the well-documented broad-spectrum anticancer activity of TNFalpha with the proven clinical usefulness of radiotherapy. Systemic delivery of the TNFalpha protein has had limited success clinically because of severe dose-limiting toxic effects. This limitation has been overcome by the use of a gene delivery approach, combined with a radiation-inducible promoter to express the TNFalpha protein in the irradiated tumour tissue. Preclinical and early phase I clinical testing indicates that effective concentrations of TNFalpha can be delivered to the tumour site without significant systemic exposure or toxic effects. The combination of radiation and TNFalpha gene delivery has produced striking antitumour effects in model systems in animals. In the clinical setting, potent anticancer activity has been observed with a high rate of complete and partial objective tumour responses. A novel mechanism of destruction of the tumour vasculature seems to be central to this distinct antitumour activity. This review summarises the rationale, mechanistic basis, preclinical data, and preliminary clinical findings for this new treatment model.
Collapse
Affiliation(s)
- Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Bromberg JS, Boros P, Ding Y, Fu S, Ku T, Qin L, Sung R. Gene transfer methods for transplantation. Methods Enzymol 2002; 346:199-224. [PMID: 11883069 DOI: 10.1016/s0076-6879(02)46057-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- J S Bromberg
- Institute for Gene Therapy and Molecular Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Smith RC, Machluf M, Bromley P, Atala A, Walsh K. Spatial and temporal control of transgene expression through ultrasound-mediated induction of the heat shock protein 70B promoter in vivo. Hum Gene Ther 2002; 13:697-706. [PMID: 11936969 DOI: 10.1089/104303402317322267] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adenoviral vectors have been constructed that express the transgenes luciferase (Adeno-HSP-Luc) or Fas ligand (Adeno-HSP-FasL) under the control of the heat shock protein 70B (hsp70B) promoter. Cultures infected with Adeno-HSP-Luc transiently expressed high levels of luciferase after heat shock. When cultures infected with Adeno-HSP-FasL were maintained at 37 degrees C, no transgene expression was observed, but when cultures were exposed to heat stress, transgene expression resulted in apoptotic cell death. In vivo, transgene expression was induced by ultrasound-mediated heating of adenovirus-infected tissue. In mice or rats injected with the Adeno-HSP-Luc construct, high levels of localized expression of luciferase activity were observed in regions subjected to ultrasound-mediated irradiation. Adeno-HSP-FasL was administered systemically to mice via the tail vein to evaluate safety. Animals receiving Adeno-HSP-FasL in the absence of ultrasound treatment did not display liver toxicity, whereas animals receiving ultrasound treatment to induce the expression of Fas ligand from the hsp70B promoter had significant increases in serum levels of liver enzymes. These data demonstrate that combining the inducible hsp70B promoter with ultrasound induction allows safe local expression of cytotoxic genes with possible therapeutic utility.
Collapse
Affiliation(s)
- Roy C Smith
- Molecular Cardiology/CVI, Whitaker Cardiovascular Institute, Boston University School of Medicine, 75 Albany Street, W611, Boston, MA 02118, USA.
| | | | | | | | | |
Collapse
|
35
|
Abstract
Parkinson's disease is one of the most likely neurological disorders to be fully treatable by drugs and new therapeutic modalities. The age-dependent and multifactorial nature of its pathogenesis allows for many strategies of intervention and repair. Most data indicate that the selectively vulnerable dopaminergic neurons in the substantia nigra of patients that have developed Parkinson's disease can be modified by protective and reparative therapies. First, the oxidative stress, protein abnormalities, and cellular inclusions typically seen could be dealt with by anti-oxidants, trophic factors, and proteolytic enhancements. Secondly, if the delay of degeneration is not sufficient, then immature dopamine neurons can be placed in the parkinsonian brain by transplantation. Such neurons can be derived from stem cell sources or even stimulated to repair from endogenous stem cells. Novel molecular and cellular treatments provide new tools to prevent and alleviate Parkinson's disease.
Collapse
Affiliation(s)
- Ole Isacson
- Neuroregeneration Laboratories, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA.
| |
Collapse
|
36
|
Kaminski JM, Nguyen K, Buyyounouski M, Pollack A. Prostate cancer gene therapy and the role of radiation. Cancer Treat Rev 2002; 28:49-64. [PMID: 12027414 DOI: 10.1053/ctrv.2002.0250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Even though prostate cancer is detected earlier than in the pre-PSA era, prostate cancer is the second leading cause of cancer mortality in the American male. Prostate cancer therapy is not ideal, especially for high-risk localized and metastatic cancer; therefore, investigators have sought new therapeutic modalities such as angiogenesis inhibitors, inhibitors of the cell signaling pathway, vaccines, and gene therapy. Gene therapy has emerged as potential therapy for both localized and systemic prostate cancer. Gene therapy has been shown to work supra-additively with radiation in controlling prostate cancer in vivo. With further technological advances in radiation therapy, gene therapy, and the understanding of prostate cancer biology, gene therapy will potentially have an important role in prostate cancer therapy.
Collapse
Affiliation(s)
- J M Kaminski
- Department of Radiation Oncology, Fox Chase Cancer Center, 7701 Burolme Avenue, Philadelphia, PA 19111, USA.
| | | | | | | |
Collapse
|
37
|
Meyer RG, Küpper JH, Kandolf R, Rodemann HP. Early growth response-1 gene (Egr-1) promoter induction by ionizing radiation in U87 malignant glioma cells in vitro. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:337-46. [PMID: 11784328 DOI: 10.1046/j.0014-2956.2001.02658.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The promoter of the early growth response gene (Egr-1) has been described to be activated by ionizing radiation, and it seems to be clear that this process involves different mitogen activated protein (MAP) kinases, dependent on the specific cell type examined. However, early steps leading to activation of the corresponding pathways and thus to overexpression of Egr-1 are not well understood. In this study, deletion mutants of the 5' upstream region of the Egr-1 gene were generated which allowed us to correlate the radiation-induction of the Egr-1 promoter in U87 glioma cells to five serum response elements. Based on the data shown, a possible role of two cAMP responsive elements for radiation-dependent promoter regulation could be ruled out. On the basis of activator/inhibitor studies applying fetal bovine serum, EGF, PD98059, anisomycin, SB203580, forskolin and wortmannin, it could be demonstrated that in U87 cells the ERK1/2 and potentially SAPK/JNK, but not the p38MAPK/SAPK2, pathway contribute to the radiation-induction of Egr-1 promoter. In addition, it was observed that irradiated cells secrete a diffusible factor into the culture media which accounts for the radiation-induced promoter upregulation. By blocking growth factor receptor activation with suramin, this effect could be completely abolished.
Collapse
Affiliation(s)
- Ralph G Meyer
- Section of Radiobiology and Molecular Environmental Research, Department of Radiotherapy, University of Tübingen, Germany
| | | | | | | |
Collapse
|
38
|
Mizuguchi H, Hayakawa T. Characteristics of adenovirus-mediated tetracycline-controllable expression system. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1568:21-9. [PMID: 11731081 DOI: 10.1016/s0304-4165(01)00195-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The combination of recombinant adenovirus (Ad) vectors and the tetracycline-controllable expression system is clearly an advantage in gene therapy and gene transfer experiment. In this study, we examined the characteristics of Ad vectors containing the tet-off or tet-on system. The Ad vector containing the tet-off system showed tightly regulatable transgene expression even at low MOI (multiplicity of infection). In contrast, regulation of gene expression by the Ad vector containing the tet-on system was not tight at low MOI, while it showed moderate regulation at high MOI (MOI=100). The Ad vector-mediated tet-on system showed lower inducible and higher background (basal) luciferase production than that of the Ad vector-mediated tet-off system. Moreover, the former system required a concentration of doxycycline, a derivative of tetracycline, approx. 2-3 log orders higher than that of the latter system to switch the luciferase expression. A combination of the vector containing the tet-on system and the vector containing the tetracycline-controlled transcriptional silencer (tTS) gene reduced the background luciferase production and improved regulation. These results suggest that the Ad vector containing the tet-off system is considered to be functionally superior to the vector containing the tet-on system. Care should be taken regarding regulation (especially lower inducibility and higher background), which is decreased in the Ad vector-mediated tet-on system in comparison with the tet-off system. The Ad vector containing the tetracycline-controllable expression system should offer a powerful tool for gene therapy and gene transfer experimentation.
Collapse
Affiliation(s)
- H Mizuguchi
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan.
| | | |
Collapse
|
39
|
Abstract
Direct targeting of cancer cells with gene therapy has the potential to treat cancer on the basis of its molecular characteristics. But although laboratory results have been extremely encouraging, many practical obstacles need to be overcome before gene therapy can fulfil its goals in the clinic. These issues are not trivial, but seem less formidable than the challenge of killing cancers selectively and rationally--a challenge that has been successfully addressed.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Biotransformation/genetics
- Bystander Effect
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Defective Viruses/genetics
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Enzymes/genetics
- Enzymes/metabolism
- Female
- Forecasting
- Gene Expression Regulation, Neoplastic/genetics
- Genes, BRCA1
- Genes, Tumor Suppressor
- Genes, p53
- Genetic Therapy/trends
- Genetic Vectors/genetics
- Genetic Vectors/immunology
- Genetic Vectors/physiology
- Genetic Vectors/therapeutic use
- Herpesviridae/genetics
- Herpesviridae/immunology
- Herpesviridae/physiology
- Humans
- Mastadenovirus/genetics
- Mastadenovirus/physiology
- Mice
- Mice, Knockout
- Mice, Transgenic
- Neoplasms/genetics
- Neoplasms/therapy
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/therapy
- Oncogenes
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/therapy
- Prodrugs/pharmacokinetics
- Prodrugs/therapeutic use
- Virus Replication
Collapse
Affiliation(s)
- F McCormick
- University of California San Francisco, Cancer Research Institute, 94115, USA.
| |
Collapse
|
40
|
Kaminski JM, Kaminski RJ, Dicker AP, Urbain JL. Defining a future role for radiogenic therapy. Cancer Treat Rev 2001; 27:289-94. [PMID: 11871865 DOI: 10.1053/ctrv.2001.0236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The goal of cancer therapy is to eliminate the cancer and/or to arrest further growth while decreasing normal tissue toxicity, i.e. to increase the therapeutic ratio. This review focuses on a group of therapeutics that are either (1) directly stimulated by radiation to produce either directly or indirectly cytotoxic agents (i.e. genes under the control of a radiation inducible promoter that produce a cytotoxic protein or an enzyme that converts a prodrug to an active form, respectively); (2) auger-electron emitting radiolabelled oligonucleotides, antibodies, nucleotide analogues, or other small molecules that are internalized; (3) radiation inducible genes that produce a ligand or transporter (or the like) which then can be targeted by cytotoxic agents (e.g. radiolabelled substance). We have termed this group of therapeutics radiogenic therapy.
Collapse
Affiliation(s)
- J M Kaminski
- Department of Radiology, Medical College of Georgia, Augusta 30912, USA.
| | | | | | | |
Collapse
|
41
|
Ito A, Shinkai M, Honda H, Kobayashi T. Heat-inducible TNF-alpha gene therapy combined with hyperthermia using magnetic nanoparticles as a novel tumor-targeted therapy. Cancer Gene Ther 2001; 8:649-54. [PMID: 11593333 DOI: 10.1038/sj.cgt.7700357] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2001] [Indexed: 11/08/2022]
Abstract
Heat-induced therapeutic gene expression is highly desired for gene therapy to minimize side effects. Furthermore, if the gene expression is triggered by heat stress, combined therapeutic effects of hyperthermia and gene therapy may be possible. We combined TNF-alpha gene therapy driven by the stress-inducible promoter, gadd 153, with hyperthermia using magnetite cationic liposomes (MCLs). In nude mice, MCLs induced cell death throughout much of the tumor area on heating under an alternating magnetic field. This heat stress also resulted in a 3-fold increase in TNF-alpha gene expression driven by the gadd 153 promoter as compared with that of nonheated tumor. TNF-alpha gene expression was also observed in the peripheral area where the hyperthermic effect was not enough to cause cell death. The combined treatment strongly arrested tumor growth in nude mice over a 30-day period, suggesting potential for cancer treatment.
Collapse
Affiliation(s)
- A Ito
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | | | | | | |
Collapse
|
42
|
Nuyts S, Van Mellaert L, Theys J, Landuyt W, Bosmans E, Anné J, Lambin P. Radio-responsive recA promoter significantly increases TNFalpha production in recombinant clostridia after 2 Gy irradiation. Gene Ther 2001; 8:1197-201. [PMID: 11509951 DOI: 10.1038/sj.gt.3301499] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2001] [Accepted: 05/23/2001] [Indexed: 11/09/2022]
Abstract
One of the major problems with gene therapy today is the lack of tumour specificity. The use of anaerobic apathogenic clostridia as a gene transfer system can target anoxic areas within the tumour. These bacteria can be genetically modified to express therapeutic proteins such as TNFalpha locally in the tumour. As shown in our results, ionising irradiation can be used in clostridia to activate genes encoding cytotoxic agents under control of a radiation-inducible promoter. A 44% significant increase (P < 0.05) in TNFalpha secretion was seen 3.5 h after a single dose of 2 Gy. A second dose of 2 Gy was also capable of repeating gene activation and gave a significant increase of TNFalpha production of 42% (P < 0.05). These results provide evidence that spatial and temporal control of gene expression can be achieved using a radio-inducible promoter. Repetitive gene activation was feasible with a second dose of 2 Gy, indicating that fractionated radiotherapy could lead to repeated gene induction resulting in prolonged and enhanced protein expression. Gene targeting by ionising radiation could thus provide a new means of increasing the therapeutic ratio in cancer treatment.
Collapse
Affiliation(s)
- S Nuyts
- Laboratory of Bacteriology, Rega Institute, Katholieke Universiteit Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
43
|
Nuyts S, Van Mellaert L, Theys J, Landuyt W, Lambin P, Anné J. The use of radiation-induced bacterial promoters in anaerobic conditions: a means to control gene expression in clostridium-mediated therapy for cancer. Radiat Res 2001; 155:716-23. [PMID: 11302769 DOI: 10.1667/0033-7587(2001)155[0716:tuorib]2.0.co;2] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Nuyts, S., Van Mellaert, L., Theys, J., Landuyt, W., Lambin, P. and Anné, J. The Use of Radiation-Induced Bacterial Promoters in Anaerobic Conditions: A Means to Control Gene Expression in Clostridium-Mediated Therapy for Cancer. Radiat. Res. 155, 716-723 (2001). Apathogenic clostridia, which have been genetically engineered to express therapeutic genes, will specifically target hypoxic and necrotic regions in tumors. This specificity can be improved further if the expression of these genes is controlled by a radiation-induced promoter, leading to spatial and temporal control of gene expression. We isolated two radiation-inducible genes of the SOS repair system of Clostridium. Northern blot experiments confirmed radiation activation of the recA and recN genes at a dose of 2 Gy. The promoter region of these genes was isolated and used to regulate expression of the lacZ gene under anaerobic conditions. For the recA promoter, a significant increase of beta-galactosidase activity of 20-30% was seen after 2 Gy irradiation. The recN promoter did not show a significant induction and had a 50-100 times lower basal expression. Treatment of the recombinant clostridial cultures with the cytostatic agent mitomycin C also resulted in a significant increase of beta-galactosidase activity that was under the control of recA or recN promoter. Oxygen does not appear to be necessary in the activation of the SOS repair system by irradiation as tested with Escherichia coli since recA-deficient and recA-containing strains showed similar survival after treatment with UV and ionizing radiation in the presence or absence of oxygen.
Collapse
Affiliation(s)
- S Nuyts
- Department of Experimental Radiobiology/Bacteriology, Laboratory of Bacteriology, Rega Institute, Katholieke Universiteit Leuven, Minderbroedersstraat 10, 3000 Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
44
|
Harrington KJ, Linardakis E, Vile RG. Transcriptional control: an essential component of cancer gene therapy strategies? Adv Drug Deliv Rev 2000; 44:167-84. [PMID: 11072113 DOI: 10.1016/s0169-409x(00)00093-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The therapeutic index of cancer gene therapy approaches will, at least in part, be dictated by the spatial and temporal control of expression of the therapeutic transgenes. Strategies which allow precise control of gene transcription are likely to play a crucial role in the future pre-clinical and clinical development of gene therapy. In this review, we discuss these issues as they relate to tissue and tumor specific promoters. In addition, the exciting opportunities offered by the development of regulated gene expression systems using small molecules, radiation and heat are reviewed. It is realistic to expect that the future offers the prospect of amalgamating elements of a number of these different systems in a co-ordinated gene delivery approach with the potential to increase the efficacy and reduce the toxicity of treatment.
Collapse
Affiliation(s)
- K J Harrington
- Molecular Medicine Program, Guggenheim 1836, Mayo Clinic, 200 1st Street SW, Rochester, MN 55902, USA
| | | | | |
Collapse
|
45
|
Abstract
Over the last 2 years, several advances have been made in the field of radiotherapy for brain tumors. Key advances are summarized in this review. Crucial technologic advances, such as radiosurgery, fractionated stereotactic radiotherapy, and intensity-modulated radiotherapy, are discussed. Better understanding of the interaction between the processes of angiogenesis, apoptosis, cell-cycle regulation, and signal transduction and the effects of ionizing radiation has made it clear that many of these "new agents" are, in fact, valuable modulators of the radiation response. Another exciting molecular discovery is the recognition of radiation-induced promoters that can be exploited to cause spatially and temporally configured expression of selected genes; this approach may represent the ideal application of conformal radiation techniques in the future, yielding well-defined genetic changes in specifically targeted tissues. The final "frontier" covered in this review is the newer categories of radiosensitizers, ranging from topoisomerase-I inhibitors, to expanded metalloporphyrins, to oxygen- dissociating agents.
Collapse
Affiliation(s)
- M P Mehta
- Department of Human Oncology, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA.
| | | | | |
Collapse
|
46
|
Manome Y, Nakamura M, Ohno T, Furuhata H. Ultrasound facilitates transduction of naked plasmid DNA into colon carcinoma cells in vitro and in vivo. Hum Gene Ther 2000; 11:1521-8. [PMID: 10945766 DOI: 10.1089/10430340050083252] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
One approach to improve the efficacy of in vivo gene therapy, with the aim at enhancing expression of the transgene, involves utilization of mechanical forces to facilitate transduction of DNA into cells. In this study, we evaluated the feasibility of mechanical insonation in gene transfers with naked DNA plasmid loading both in vitro and in vivo. We used an ultrasound probe, which can focus the ultrasonic beam in the exit zone of the probe. The reporter pcDNA3-lacZ plasmid, containing Escherichia coli lacZ or the beta-galactosidase gene (beta-gal), and the neomycin 3'-phosphotransferase gene (neo), was used for evaluation of transfer efficiency. Expression of beta-gal in MC38 murine colon carcinoma cells was measured after insonation of 20 W/cm2 with continuous 1.0-MHz wave exposure. In a transient assay, significant numbers of cells were transduced with the beta-galactosidase gene. After cells were treated with geneticin, we also observed a difference in colonogenicity between noninsonated and insonated groups. When MC38 cells were implanted in syngeneic mice and plasmid was injected, the insonation that followed facilitated beta-galactosidase expression. These results indicate that insonation represents a potential approach for gene therapy when combined with naked DNA plasmid injection.
Collapse
Affiliation(s)
- Y Manome
- Department of Microbiology, Jikei University School of Medicine, Tokyo, Japan
| | | | | | | |
Collapse
|
47
|
Scott SD, Marples B, Hendry JH, Lashford LS, Embleton MJ, Hunter RD, Howell A, Margison GP. A radiation-controlled molecular switch for use in gene therapy of cancer. Gene Ther 2000; 7:1121-5. [PMID: 10918478 DOI: 10.1038/sj.gt.3301223] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ionising radiation induces the expression of a number of radiation-responsive genes and there is current interest in exploiting this to regulate the expression of exogenous therapeutic genes in gene therapy strategies for cancer. However, the radiation-responsive promoters used in these approaches are often associated with low and transient levels of therapeutic gene expression. We describe here a novel radiation-triggered molecular switching device based on promoter elements from the radiation-responsive Egr-1 gene and the cre-LoxP site-specific recombination system of the P1 bacteriophage. Using this system, a single, minimally toxic dose of radiation induced cre-mediated excision of a lox-P flanked stop cassette in a silenced expression vector and this resulted in amplified levels of CMV-promoter-driven expression of the exogenous tumour-sensitising gene, HSV-tk. This strategy could be used in combination with targeted delivery and tumour-specific promoters to elicit the tumour-targeted and prolonged expression of a variety of tumour-sensitising genes and provide an unprecedented level of control and tumour selectivity.
Collapse
Affiliation(s)
- S D Scott
- Cancer Research Campaign Section of Genome Damage and Repair, Paterson Institute for Cancer Research, Christie Hospital (NHS) Trust, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Worthington J, Robson T, Murray M, O'Rourke M, Keilty G, Hirst DG. Modification of vascular tone using iNOS under the control of a radiation-inducible promoter. Gene Ther 2000; 7:1126-31. [PMID: 10918479 DOI: 10.1038/sj.gt.3301224] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
It may be therapeutically advantageous to alter tumour blood supply specifically. Nitric oxide is a potent vasodilator which is produced in many tissues by the enzyme nitric oxide synthase (NOS). We have transfected cDNA for the inducible isoform of this enzyme (iNOS), under the control of the radiation-inducible promoter WAF1. The activity of the promoter was initially assessed using green fluorescent protein (GFP) in both endothelial cells and rat tail artery segments. Induction of protein expression by 9.5- and 4.5-fold respectively, was observed after a radiation dose of 4 Gy. Artery sections were then transfected with the WAF1/iNOS construct; this gave five-fold induction of iNOS protein after a dose of 4 Gy. The transfected artery was also tested functionally for relaxation, indicative of NO production. One hour after exposure to 4 Gy there was a significant (65%) relaxation of artery segments that had been preconstricted with phenylephrine. This could be partially reversed by the NOS inhibitor nitro-L-arginine. This study demonstrates that we can regulate vascular tone using an X-ray inducible promoter.
Collapse
Affiliation(s)
- J Worthington
- Radiation Science Research Group, School of Biomedical Sciences, University of Ulster, Newtownabbey, Co Antrim, UK
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
One of the biggest challenges facing cancer therapy is to generate tumour-specific treatment strategies. Gene therapy hopes to achieve this by directing the activity of therapeutic genes specifically to the sites of disease. Of paramount importance for the success of this approach is the availability of tumour-specific delivery systems: both the transductional targeting of the vector vehicle and the restriction of transgene expression to the tumour are promising strategies towards this goal. This review will focus on the recent achievements in the field of transcriptional targeting and the different strategies to improve or design promoters with the desired specificities.
Collapse
Affiliation(s)
- D M Nettelbeck
- Institute of Molecular Biology and Tumor Research (IMT), Philipps-University Marburg, Emil-Mannkopff-Strasse 2, D-35033 Marburg, Germany.
| | | | | |
Collapse
|
50
|
Vekris A, Maurange C, Moonen C, Mazurier F, De Verneuil H, Canioni P, Voisin P. Control of transgene expression using local hyperthermia in combination with a heat-sensitive promoter. J Gene Med 2000; 2:89-96. [PMID: 10809142 DOI: 10.1002/(sici)1521-2254(200003/04)2:2<89::aid-jgm90>3.0.co;2-j] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Local production of therapeutic proteins, e.g. for cancer treatments, is based on gene therapy approaches and requires tight spatial and temporal control of gene expression. Here we demonstrate the use of local hyperthermia of varying intensity and duration to control the expression of a transgene under control of the thermoinducible hsp70 (heat shock protein) promoter. METHODS Heat-induced expression of the EGFP (green fluorescent protein) reporter gene was characterized using a stably transfected glioma C6 cell line expressing the EGFP gene under control of the heat inducible minimal hsp70 promoter both in vitro and in vivo for subcutaneous tumors in immunodeficient mice. RESULTS A heat shock of 20-30 min at 43 degrees C in cell culture led to a maximum EGFP concentration at about 24 h. Heat treatments at higher temperature (up to 48 degrees C) but with shorter durations (down to 30 s) also induced strong EGFP expression. Local heating in situ led to gradients in EGFP expression which decreased with increasing distance from the heat source. CONCLUSION Local hyperthermia, in combination with a heat sensitive promoter, represents a method for the spatial and temporal control of transgene expression.
Collapse
Affiliation(s)
- A Vekris
- Laboratoire de Resonance Magnétique des Sysèmes Biologiques, UMR5536 CNRS-Université V. Segalen, Bordeaux, France
| | | | | | | | | | | | | |
Collapse
|