Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Publication year range
1.
Mol Ther ; 31(10): 2839-2860, 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37574780

ABSTRACT

Gliomas are the most prevalent and devastating primary malignant brain tumors in adults. Despite substantial advances in understanding glioma biology, there have been no regulatory drug approvals in the US since bevacizumab in 2009 and tumor treating fields in 2011. Recent phase III clinical trials have failed to meet their prespecified therapeutic primary endpoints, highlighting the need for novel therapies. The poor prognosis of glioma patients, resistance to chemo-radiotherapy, and the immunosuppressive tumor microenvironment underscore the need for the development of novel therapies. Gene therapy-based immunotherapeutic strategies that couple the ability of the host immune system to specifically kill glioma cells and develop immunological memory have shown remarkable progress. Two adenoviral vectors expressing Ad-HSV1-TK/GCV and Ad-Flt3L have shown promising preclinical data, leading to FDA approval of a non-randomized, phase I open-label, first in human trial to test safety, cytotoxicity, and immune-stimulatory efficiency in high-grade glioma patients (NCT01811992). This review provides a thorough overview of immune-stimulatory gene therapy highlighting recent advancements, potential drawbacks, future directions, and recommendations for future implementation of clinical trials.


Subject(s)
Brain Neoplasms , Glioma , Animals , Humans , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Rodentia/genetics , Adenoviridae/genetics , Glioma/genetics , Glioma/therapy , Glioma/pathology , Genetic Therapy , Thymidine Kinase/genetics , Genetic Vectors/genetics , Tumor Microenvironment
2.
Biotechnol Appl Biochem ; 71(1): 5-16, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37743549

ABSTRACT

Suicide gene therapy involves introducing viral or bacterial genes into tumor cells, which enables the conversion of a nontoxic prodrug into a toxic-lethal drug. The application of the bacterial cytosine deaminase (bCD)/5-fluorocytosine (5-FC) approach has been beneficial and progressive within the current field of cancer therapy because of the enhanced bystander effect. The basis of this method is the preferential deamination of 5-FC to 5-fluorouracil by cancer cells expressing cytosine deaminase (CD), which strongly inhibits DNA synthesis and RNA function, effectively targeting tumor cells. However, the poor binding affinity of toward 5-FC compared to the natural substrate cytosine and/or inappropriate thermostability limits the clinical applications of this gene therapy approach. Nowadays, many genetic engineering studies have been carried out to solve and improve the activity of this enzyme. In the current review, we intend to discuss the biotechnological aspects of Escherichia coli CD, including its structure, functions, molecular cloning, and protein engineering. We will also explore its relevance in cancer clinical trials. By examining these aspects, we hope to provide a thorough understanding of E. coli CD and its potential applications in cancer therapy.


Subject(s)
Cytosine Deaminase , Prodrugs , Humans , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Escherichia coli/metabolism , Fluorouracil/chemistry , Flucytosine/pharmacology , Flucytosine/metabolism , Genetic Therapy , Prodrugs/metabolism
3.
Int J Mol Sci ; 25(5)2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38473904

ABSTRACT

Cancer in dogs has increased in recent years and is a leading cause of death. We have developed a retroviral replicating vector (RRV) that specifically targets cancer cells for infection and replication. RRV carrying a suicide gene induced synchronized killing of cancer cells when administered with a prodrug after infection. In this study, we evaluated two distinct RRVs derived from amphotropic murine leukemia virus (AMLV) and gibbon ape leukemia virus (GALV) in canine tumor models both in vitro and in vivo. Despite low infection rates in normal canine cells, both RRVs efficiently infected and replicated within all the canine tumor cells tested. The efficient intratumoral spread of the RRVs after their intratumoral injection was also demonstrated in nude mouse models of subcutaneous canine tumor xenografts. When both RRVs encoded a yeast cytosine deaminase suicide gene, which converts the prodrug 5-fluorocytosine (5-FC) to the active drug 5-fluorouracil, they caused tumor-cell-specific 5-FC-induced killing of the canine tumor cells in vitro. Furthermore, in the AZACF- and AZACH-cell subcutaneous tumor xenograft models, both RRVs exerted significant antitumor effects. These results suggest that RRV-mediated suicide gene therapy is a novel therapeutic approach to canine cancers.


Subject(s)
Neoplasms , Prodrugs , Mice , Humans , Dogs , Animals , Genetic Therapy/methods , Cell Line, Tumor , Leukemia Virus, Gibbon Ape/genetics , Fluorouracil/pharmacology , Flucytosine/pharmacology , Prodrugs/pharmacology , Genetic Vectors , Cytosine Deaminase/genetics , Neoplasms/drug therapy
4.
Biochem Biophys Res Commun ; 666: 107-114, 2023 07 23.
Article in English | MEDLINE | ID: mdl-37182285

ABSTRACT

Cellular behavior is heavily influenced by cellular interactions, which are often lost in conventional cell culture methods. As a result, in vitro cellular behavior may not accurately reflect in vivo conditions. Three-dimensional (3D) culture, on the other hand, is better suited for studying cellular behavior as it allows for more comprehensive cell communication. In this study, we utilized 3D culture of the MCF-7 cell line to create a minimal model of luminal-A breast cancer and evaluated its histopathological and morphological features using various methods. To determine the optimal therapeutic strategies for eliminating cancer cells, we assessed the effectiveness of diverse therapeutic approaches, including targeting distinct phases of the cell cycle, endocrine therapy, and gene therapy in both 2D and 3D culture systems. Our findings indicate that cells derived from mammospheres respond differently to their parent cells in monolayer culture depending on the therapeutic strategy used. This variability in drug response may be due to the altered microenvironment created by heterogeneous cellular makeup and emerging cellular interactions in the 3D culture. Therefore, it is important to administer a therapeutic approach that can eradicate cells regardless of the microenvironment.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , MCF-7 Cells , Cell Culture Techniques/methods , Cell Communication , Cell Line, Tumor , Tumor Microenvironment
5.
J Nanobiotechnology ; 20(1): 340, 2022 Jul 20.
Article in English | MEDLINE | ID: mdl-35858873

ABSTRACT

Practice of tumor-targeted suicide gene therapy is hampered by unsafe and low efficient delivery of plasmid DNA (pDNA). Using HIV-Tat-derived peptide (Tat) to non-covalently form Tat/pDNA complexes advances the delivery performance. However, this innovative approach is still limited by intracellular delivery efficiency and cell-cycle status. In this study, Tat/pDNA complexes were further condensed into smaller, nontoxic nanoparticles by Ca2+ addition. Formulated Tat/pDNA-Ca2+ nanoparticles mainly use macropinocytosis for intercellular delivery, and their macropinocytic uptake was persisted in mitosis (M-) phase and highly activated in DNA synthesis (S-) phase of cell-cycle. Over-expression or phosphorylation of a mitochondrial chaperone, 75-kDa glucose-regulated protein (GRP75), promoted monopolar spindle kinase 1 (MPS1)-controlled centrosome duplication and cell-cycle progress, but also driven cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles. Further in vivo molecular imaging based on DF (Fluc-eGFP)-TF (RFP-Rluc-HSV-ttk) system showed that Tat/pDNA-Ca2+ nanoparticles exhibited highly suicide gene therapy efficiency in mouse model xenografted with human ovarian cancer. Furthermore, arresting cell-cycle at S-phase markedly enhanced delivery performance of Tat/pDNA-Ca2+ nanoparticles, whereas targeting GRP75 reduced their macropinocytic delivery. More importantly, in vivo targeting GRP75 combined with cell-cycle or macropinocytosis inhibitors exhibited distinct suicide gene therapy efficiency. In summary, our data highlight that mitochondrial chaperone GRP75 moonlights as a biphasic driver underlying cell-cycle-dependent macropinocytosis of Tat/pDNA-Ca2+ nanoparticles in ovarian cancer.


Subject(s)
Nanoparticles , Ovarian Neoplasms , Animals , Calcium , DNA/chemistry , Female , Gene Transfer Techniques , Genetic Therapy , HSP70 Heat-Shock Proteins , Humans , Membrane Proteins , Mice , Nanoparticles/chemistry , Ovarian Neoplasms/therapy , Plasmids , Transfection
6.
Molecules ; 27(23)2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36500454

ABSTRACT

Uterine leiomyoma is the most common benign tumor of the reproductive system. Current therapeutic options do not simultaneously meet the requirements of long-term efficiency and fertility preservation. Suicide gene delivery can be proposed as a novel approach to uterine leiomyoma therapy. Non-viral vehicles are an attractive approach to DNA delivery for gene therapy of both malignant and benign tumors. Peptide-based vectors are among the most promising candidates for the development of artificial viruses, being able to efficiently cross barriers of DNA transport to cells. Here we described nanoparticles composed of cysteine-crosslinked polymer and histidine-arginine-rich peptide modified with iRGD moiety and characterized them as vehicles for plasmid DNA delivery to pancreatic cancer PANC-1 cells and the uterine leiomyoma cell model. Several variants of nanoparticles were formulated with different targeting ligand content. The physicochemical properties that were studied included DNA binding and protection, interaction with polyanions and reducing agents, size, structure and zeta-potential of the peptide-based nanoparticles. Cytotoxicity, cell uptake and gene transfection efficiency were assessed in PANC-1 cells with GFP and LacZ-encoding plasmids. The specificity of gene transfection via αvß3 integrin binding was proved in competitive transfection. The therapeutic potential was evaluated in a uterine leiomyoma cell model using the suicide gene therapy approach. The optimal formulation was found to be at the polyplex with the highest iRGD moiety content being able to transfect cells more efficiently than control PEI. Suicide gene therapy using the best formulation resulted in a significant decrease of uterine leiomyoma cells after ganciclovir treatment. It can be concluded that the application of iRGD-modified peptide-based nanoparticles has a high potential for cellular delivery of DNA therapeutics in favor of uterine leiomyoma gene therapy.


Subject(s)
Nanoparticles , Neoplasms , Humans , Integrins/genetics , Transfection , Peptides/chemistry , Nanoparticles/chemistry , DNA/chemistry , Plasmids
7.
Neurosurg Rev ; 44(1): 29-49, 2021 Feb.
Article in English | MEDLINE | ID: mdl-31781985

ABSTRACT

Malignant glioma, which is characterized by diffuse infiltration into the normal brain parenchyma, is the most aggressive primary brain tumor with dismal prognosis. Over the past 40 years, the median survival has only slightly improved. Therefore, new therapeutic modalities must be developed. In the 1990s, suicide gene therapy began attracting attention for the treatment of malignant glioma. Some clinical trials used a viral vector for suicide gene transduction; however, it was found that viral vectors cannot cover the large invaded area of glioma cells. Interest in this therapy was recently revived because some types of stem cells possess a tumor-tropic migratory capacity, which can be used as cellular delivery vehicles. Immortalized, clonal neural stem cell (NSC) line has been used for patients with recurrent high-grade glioma, which showed safety and efficacy. Embryonic and induced pluripotent stem cells may be considered as sources of NSC because NSC is difficult to harvest, and ethical issues have been raised. Mesenchymal stem cells are alternative candidates for cellular vehicle and are easily harvested from the bone marrow. In addition, a new type of nonlytic, amphotropic retroviral replicating vector encoding suicide gene has shown efficacy in patients with recurrent high-grade glioma in a clinical trial. This replicating viral capacity is another possible candidate as delivery vehicle to tackle gliomas. Herein, we review the concept of suicide gene therapy, as well as recent progress in preclinical and clinical studies in this field.


Subject(s)
Brain Neoplasms/therapy , Genes, Transgenic, Suicide/genetics , Genetic Therapy/methods , Glioma/therapy , Clinical Trials as Topic , Humans
8.
Int J Mol Sci ; 22(17)2021 Aug 29.
Article in English | MEDLINE | ID: mdl-34502287

ABSTRACT

Gene-directed enzyme prodrug gene therapy (GDEPT) theoretically represents a useful method to carry out chemotherapy for cancer with minimal side effects through the formation of a chemotherapeutic agent inside cancer cells. However, despite great efforts, promising preliminary results, and a long period of time (over 25 years) since the first mention of this method, GDEPT has not yet reached the clinic. There is a growing consensus that optimal cancer therapies should generate robust tumor-specific immune responses. The advent of checkpoint immunotherapy has yielded new highly promising avenues of study in cancer therapy. For such therapy, it seems reasonable to use combinations of different immunomodulators alongside traditional methods, such as chemotherapy and radiotherapy, as well as GDEPT. In this review, we focused on non-viral gene immunotherapy systems combining the intratumoral production of toxins diffused by GDEPT and immunomodulatory molecules. Special attention was paid to the applications and mechanisms of action of the granulocyte-macrophage colony-stimulating factor (GM-CSF), a cytokine that is widely used but shows contradictory effects. Another method to enhance the formation of stable immune responses in a tumor, the use of danger signals, is also discussed. The process of dying from GDEPT cancer cells initiates danger signaling by releasing damage-associated molecular patterns (DAMPs) that exert immature dendritic cells by increasing antigen uptake, maturation, and antigen presentation to cytotoxic T-lymphocytes. We hypothesized that the combined action of this danger signal and GM-CSF issued from the same dying cancer cell within a limited space would focus on a limited pool of immature dendritic cells, thus acting synergistically and enhancing their maturation and cytotoxic T-lymphocyte attraction potential. We also discuss the problem of enhancing the cancer specificity of the combined GDEPT-GM-CSF-danger signal system by means of artificial cancer specific promoters or a modified delivery system.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Immunotherapy/methods , Neoplasms/therapy , Animals , Cancer Vaccines/pharmacology , Genes, Transgenic, Suicide , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Neoplasms/immunology , Oncolytic Virotherapy/methods , Prodrugs/pharmacology , Thymidine Kinase/genetics , Thymidine Kinase/pharmacology
9.
Cancer Cell Int ; 20: 26, 2020.
Article in English | MEDLINE | ID: mdl-32002015

ABSTRACT

BACKGROUND: Therapeutic approaches for cancer rely on careful consideration of finding the optimal way of delivering the pro-drug for cellular-based cancer treatment. Cell lines and cell cultures have been used in these studies to compare the in vitro and in vivo efficacy of autologous vs. allogeneic tumour cellular gene therapy. Here we have investigated and are reporting for the first time the effect of prodrug ganciclovir (GCV)-preloading (pre-treatment) in suicide gene therapy of cancer. METHODS: This study examines the effect of GCV-preloading (pre-treatment) on a range of tumour cell lines in conjunction with suicide gene therapy of cancer. To determine the efficacy of this modality, a series of in vitro and in vivo experiments were conducted using genetically modified and unmodified tumour cell lines. RESULTS: Following co-culture of herpes simplex virus thymidine kinase (HSV-TK) modified tumour cells and unmodified tumour cells both in vitro and in vivo, GCV-preloading (pre-treatment) of TK-modified human and mouse mesothelioma cells and ovarian tumour cells allowed them to mediate efficiently bystander killing of neighbouring unmodified tumour cells in vitro. In contrast, GCV-preloading of TK-modified human and mouse mesothelioma cells and ovarian tumour cells abolished their in vivo ability to induce bystander killing of unmodified tumour cells, although there was some tumour regression compared to control groups but this was not statistically significant. These results suggest that preloading TK modified tumour cells with GCV needs further study to define the most effective strategy for an in vivo application to retain their bystander killing potential after exposure to lethal doses of GCV in vitro. CONCLUSIONS: This study highlights the promising possibility of improving the efficacy of pro-drug system to prevent any damage to the immune system and enhancing this type of suicide gene therapy of cancer, as well as the need for further studies to explore the discrepancies between in vitro and in vivo results.

10.
Int J Hyperthermia ; 37(1): 101-109, 2020.
Article in English | MEDLINE | ID: mdl-31969028

ABSTRACT

Objective: To establish the technique of intratumoral combination therapy of radiofrequency hyperthermia (RFH) with herpes simplex virus-thymidine kinase/ganciclovir (HSV-TK/GCV) gene therapy for rat ovarian cancers.Material and methods: This study consisted of three parts: (1) in vitro experiments to establish the 'proof of principal' that combination of RFH and HSV-TK gene therapy has the synergistic effect on human ovarian cancer cells; (2) creation of bioluminescence imaging-detectable rat ovarian cancer model; and (3) in vivo experiments using this rat model to validate the technical feasibility of the combination therapy. Cells and nude rats were divided into four groups: (i) combination therapy (HSV-TK/GCV + RFH); (ii) RFH; (iii) HSV-TK/GCV; and (iv) phosphate-buffered saline (PBS). Data were analyzed using Dunnett t-test or Kruskal-Wallis test.Results: Cell proliferation assay demonstrated significantly greater reduction in viable cells with the combination therapy [0.52 (0.43, 0.61)] compared to other treatments [RFH 0.90 (0.84, 0.96), HSV-TK/GCV 0.71 (0.53, 0.88), PBS 1 (1, 1); p < .05]. For 24 rat models with bioluminescence imaging-detectable orthotopic ovarian cancer (n = 6 per group), optical imaging demonstrated significantly decreased relative bioluminescence signal with the combination therapy [0.81 (0.52, 1.08)] compared to other treatments [RFH 3.60 (2.34, 4.86), HSV-TK/GCV 2.21 (1.71, 2.71), PBS 3.74 (3.19, 4.29); p < .001]. Ultrasound imaging demonstrated the smallest relative tumor volume with the combination therapy [0.78 (0.45, 1.11) versus 3.50 (2.67, 4.33), 2.10 (0.83, 3.37), 3.70 (1.79, 5.61); p < .05].Conclusion: The feasibility of intratumoral RFH-enhanced HSV-TK/GCV gene therapy was established on a unique rat model with molecular imaging-detectable orthotopic ovarian cancer.


Subject(s)
Genetic Therapy/methods , Hyperthermia, Induced/methods , Molecular Imaging/methods , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/radiotherapy , Simplexvirus/drug effects , Thymidine Kinase/therapeutic use , Animals , Female , Humans , Rats , Rats, Nude , Thymidine Kinase/pharmacology
11.
BMC Cancer ; 19(1): 921, 2019 Sep 14.
Article in English | MEDLINE | ID: mdl-31521130

ABSTRACT

BACKGROUND: Free flap-mediated gene therapy in the tumor bed following surgical resection is a promising approach in cancer targeted treatment of residual disease. We investigated the selective killing efficacy of a lentivirus-mediated cytosine deaminase-thymidine kinase (CDglyTK) gene in transplanted breast cancer delivered into a free flap by intra-artery perfusion. METHODS: Proliferation, apoptosis, and cell cycle of rat SHZ-88 breast cancer cells transfected with a lentivirus-mediated CD/TK gene were measured following treatment with ganciclovir and 5-flucytosine in vitro. A model of residual disease of breast cancer in a rat superficial inferior epigastric artery (SIEA) flap model was used to study the therapeutic potential of a double suicide CD/TK and prodrug system in vivo. RESULTS: Killing efficacy of the double suicide CD/TK and prodrug system on SHZ-88 cells was mediated by increased apoptosis and cell cycle arrest at the G1 phase with significant bystander effect. Following recombinant lentivirus transfection of rat SIEA flap by intra-artery perfusion, CD/TK gene expression was limited to the flap, and the volume and weight of transplanted tumors were significantly reduced without observable toxicity. CONCLUSIONS: SIEA flaps transfected with a lentivirus-mediated CDglyTK gene by intra-artery perfusion effectively suppress transplanted breast tumor growth without obvious systemic toxic effects in rats.


Subject(s)
Breast Neoplasms/genetics , Cytosine Deaminase/genetics , Free Tissue Flaps , Genetic Therapy , Genetic Vectors/genetics , Lentivirus/genetics , Recombinant Fusion Proteins , Thymidine Kinase/genetics , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Bystander Effect , Cell Line, Tumor , Cell Survival/drug effects , Combined Modality Therapy , Disease Models, Animal , Female , Ganciclovir/pharmacology , Genes, Transgenic, Suicide , Genetic Vectors/administration & dosage , Humans , Perfusion , Rats , Transgenes , Treatment Outcome
12.
J Gene Med ; 20(10-11): e3054, 2018 10.
Article in English | MEDLINE | ID: mdl-30172246

ABSTRACT

BACKGROUND: Cervical cancer is the third most common cause of cancer in women. The 5-year survival rate in oropharyngeal squamous cell carcinomas is approximately 50% and this rate has not improved in recent decades. These cancers are accessible to direct intervention. We examined the ability of a highly efficient non-viral vector, TransfeX (ATCC, Manassas, VA, USA), to deliver the suicide gene HSV-tk to cervical, oral and pharyngeal cancer cells and to induce cytotoxicity following the administration of the prodrug, ganciclovir. METHODS: HeLa cervical carcinoma, HSC-3 and H357 oral squamous cell carcinoma and FaDu pharyngeal carcinoma cells were transfected with cytomegalovirus (CMV)- or enhanced episomal vector (EEV)-driven HSV-tk plasmids and treated with ganciclovir for 24-120 h. Cell viability was assessed by Alamar blue. RESULTS: The viability of HeLa cells was reduced to only 30-40%, despite the very high levels of transgene expression. By contrast, the viability of HSC-3 cells was reduced to 10%, although transgene expression was 18-fold lower than that in HeLa cells. An approximately five-fold higher transgene expression was obtained with the EEV-plasmid than from the CMV-plasmid. Nevertheless, HeLa cell viability after suicide gene + ganciclovir treatment was reduced by only 35% compared to 70% with the CMV-plasmid. For HSC-3 cells, the reduction was 40% for the EEV- and 80% for the CMV-plasmid. The lower efficiency of transfection with the EEV-plasmid may explain the lower cytotoxicity. CONCLUSIONS: TransfeX-mediated gene delivery to cervical, pharyngeal and oral cancer cells may be used for suicide gene therapy. The levels of transgene expression, however, do not translate directly to cytotoxicity.


Subject(s)
Cytomegalovirus/genetics , Gene Transfer Techniques , Genes, Transgenic, Suicide/genetics , Plasmids/genetics , Antiviral Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Female , Ganciclovir/pharmacology , Genetic Therapy/methods , Genetic Vectors/genetics , HeLa Cells , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Pharyngeal Neoplasms/genetics , Pharyngeal Neoplasms/pathology , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
13.
Mol Biol Rep ; 45(6): 2263-2273, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30242665

ABSTRACT

The potential of herpes simplex virus type 1 thymidine kinase (HSV-tk)-expressing olfactory ensheathing cells (OEC) treated with ganciclovir (GCV) to induce cell death in adjacent HSV-tk-negative cells (bystander effect) has been well demonstrated. Although it has been shown that bystander effect occurs through the delivery of phosphorylated GCV, the bystander effect mechanism and the role of gap junctions for human OECs mediated suicide gene therapy in primary astrocytes of human glioblastma remain obscure. In the present study, the efficacy of a new method for the transfer of phosphorylated GCV from OECs into primary astrocytes was evaluated. Surgical biopsy of glioblastoma was used to isolate primary astrocyte. Biopsy of olfactory mucosa was applied to isolate olfactory ensheathing cell. Expression of S100-beta antigen was confirmed immunocytochemically in astrocytes and OECs. OECs were transduced to lentiviral containing thymidine kinase gene (TK) and co-cultured with astrocytes. Fluorescent dye transfer and western blot analysis indicated the expression of connexin43 between olfactory ensheathing cells and astrocytes whereas, expression of the gap junction protein connexin43 was inhibited by the gap junction inhibitor 18α-glycyrrhethinic acid (AGA, 20 µg/ml). Furthermore, co-culture of astrocytes with OEC-TK in the presence of concentration of 30 µg/ml GCV led to a decrease in astrocytes survival rate. Also, apoptosis hallmarks, including DNA fragmentation in cell nuclear, expression increase of Bax to Bcl-2 ratio and increase of caspase3 activation were observed in this study. Our findings suggest that human olfactory ensheathing cells can deliver phosphorylated GCV into the glioblastoma derived astrocytes through gap junction communication for apoptosis induction.


Subject(s)
Astrocytes/drug effects , Ganciclovir/administration & dosage , Glioblastoma/drug therapy , Adult , Antiviral Agents , Bystander Effect , Coculture Techniques , Connexin 43/drug effects , Ganciclovir/pharmacology , Gap Junctions/drug effects , Genes, Reporter , Genetic Therapy , Genetic Vectors , Herpesvirus 1, Human , Humans , Olfactory Mucosa , Primary Cell Culture , Thymidine Kinase/therapeutic use , Tumor Cells, Cultured
14.
Mol Ther ; 25(2): 356-367, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28153088

ABSTRACT

Mutations in the KRAS gene, which persistently activate RAS function, are most frequently found in many types of human cancers. Here, we proposed and verified a new approach against cancers harboring the KRAS mutation with high cancer selectivity and efficient anti-cancer effects based on targeted RNA replacement. To this end, trans-splicing ribozymes from Tetrahymena group I intron were developed, which can specifically target and reprogram the mutant KRAS G12V transcript to induce therapeutic gene activity in cells. Adenoviral vectors containing the specific ribozymes with downstream suicide gene were constructed and then infection with the adenoviruses specifically downregulated KRAS G12V expression and killed KRAS G12V-harboring cancer cells additively upon pro-drug treatment, but it did not affect the growth of wild-type KRAS-expressing cells. Minimal liver toxicity was noted when the adenoviruses were administered systemically in vivo. Importantly, intratumoral injection of the adenoviruses with pro-drug treatment specifically and significantly impeded the growth of xenografted tumors harboring KRAS G12V through a trans-splicing reaction with the target RNA. In contrast, xenografted tumors harboring wild-type KRAS were not affected by the adenoviruses. Therefore, RNA replacement with a mutant KRAS-targeting trans-splicing ribozyme is a potentially useful therapeutic strategy to combat tumors harboring KRAS mutation.


Subject(s)
Mutation , Neoplasms/genetics , Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , RNA/genetics , Targeted Gene Repair , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Expression , Gene Order , Genetic Vectors/genetics , Humans , Male , Mice , Proto-Oncogene Proteins p21(ras)/metabolism , RNA/metabolism , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , Trans-Splicing , Xenograft Model Antitumor Assays
15.
Int J Mol Sci ; 19(1)2018 Jan 09.
Article in English | MEDLINE | ID: mdl-29315221

ABSTRACT

Since their invention in 2006, induced Pluripotent Stem (iPS) cells remain a great promise for regenerative medicine circumventing the ethical issues linked to Embryonic Stem (ES) cell research. iPS cells can be generated in a patient-specific manner as an unlimited source of various cell types for in vitro drug screening, developmental biology studies and regenerative use. Having the capacity of differentiating into the cells of all three primary germ layers, iPS cells have high potential to form teratoma tumors. This remains their main disadvantage and hazard which, until resolved, prevents utilization of iPS cells in clinic. Here, we present an approach for increasing iPS cells safety by introducing genetic modification-exogenous suicide gene Herpes Simplex Virus Thymidine Kinase (HSV-TK). Its expression results in specific vulnerability of genetically modified cells to prodrug-ganciclovir (GCV). We show that HSV-TK expressing cells can be eradicated both in vitro and in vivo with high specificity and efficiency with low doses of GCV. Described strategy increases iPS cells safety for future clinical applications by generating "emergency exit" switch allowing eradication of transplanted cells in case of their malfunction.


Subject(s)
Simplexvirus/genetics , Thymidine Kinase/metabolism , Viral Proteins/metabolism , Animals , Apoptosis , Cellular Reprogramming , Female , Ganciclovir/pharmacology , Gene Expression/drug effects , Genes, Transgenic, Suicide/genetics , Hair/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Keratinocytes/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology , Neoplasms/therapy , Simplexvirus/enzymology , Teratoma/pathology , Thymidine Kinase/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Viral Proteins/genetics
16.
Int J Mol Sci ; 19(4)2018 Mar 27.
Article in English | MEDLINE | ID: mdl-29584688

ABSTRACT

Colorectal cancer is the most common cancer in both men and women and the second most common cause of cancer-related deaths. Suicide gene-based therapy with suicide gene-transduced mesenchymal stem cells (MSCs) is a promising therapeutic strategy. A tetracycline-controlled Tet-On inducible system used to regulate gene expression may be a useful tool for gene-based therapies. The aim of this study was to develop therapeutic MSCs with a suicide gene that is induced by an artificial stimulus, to validate therapeutic gene expression, and to monitor the MSC therapy for colon cancer using optical molecular imaging. For our study, we designed the Tet-On system using a retroviral vector and developed a response plasmid RetroX-TRE (tetracycline response element) expressing a mutant form of herpes simplex virus thymidine kinase (HSV1-sr39TK) with dual reporters (eGFP-Fluc2). Bone marrow-derived MSCs were transduced using a RetroX-Tet3G (Clontech, CA, USA) regulatory plasmid and RetroX-TRE-HSV1-sr39TK-eGFP-IRES-Fluc2, for a system with a Tet-On (MSC-Tet-TK/Fluc2 or MSC-Tet-TK) or without a Tet-On (MSC-TK/Fluc2 or MSC-TK) function. Suicide gene engineered MSCs were co-cultured with colon cancer cells (CT26/Rluc) in the presence of the prodrug ganciclovir (GCV) after stimulation with or without doxycycline (DOX). Treatment efficiency was monitored by assessing Rluc (CT26/Rluc) and Fluc (MSC-Tet-TK and MSC-TK) activity using optical imaging. The bystander effect of therapeutic MSCs was confirmed in CT26/Rluc cells after GCV treatment. Rluc activity in CT26/Rluc cells decreased significantly with GCV treatment of DOX(+) cells (p < 0.05 and 0.01) whereas no significant changes were observed in DOX(-) cells. In addition, Fluc activity in also decreased significantly with DOX(+) MSC-Tet-TK cells, but no signal was observed in DOX(-) cells. In addition, an MSC-TK bystander effect was also confirmed. We assessed therapy with this system in a colon cancer xenograft model (CT26/Rluc). We successfully transduced cells and developed a Tet-On system with the suicide gene HSV1-sr39TK. Our results confirmed the therapeutic efficiency of a suicide gene with the Tet-On system for colon cancer. In addition, our results provide an innovative therapeutic approach using the Tet-On system to eradicate tumors by administration of MSC-Tet-TK cells with DOX and GCV.


Subject(s)
Colonic Neoplasms/therapy , Genes, Transgenic, Suicide , Mesenchymal Stem Cells/cytology , Molecular Imaging/methods , Animals , Apoptosis , Bystander Effect , Cell Line, Tumor , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/genetics , Doxycycline/pharmacology , Female , Ganciclovir/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Mice , Optical Imaging , Transduction, Genetic , Xenograft Model Antitumor Assays
17.
J Gene Med ; 19(8)2017 Aug.
Article in English | MEDLINE | ID: mdl-28667693

ABSTRACT

BACKGROUND: Lyophilization is an effective method for preserving nonviral gene vectors. To improve the stability and transgene expression of lyophilized plasmid DNA (pDNA) complexes, we coated the surfaces of pDNA/chitosan complexes with hyaluronic acid (HA) of varying molecular masses. The transgene expression of pDNA/chitosan/HA ternary complexes was characterized in vitro and in vivo. METHODS: pDNA complexes were lyophilized overnight and the resultant products with spongy, porous consistencies were stored at -30, 4 or 25°C for 2 weeks. Rehydrated complexes were characterized using gel retardation assays, aiming to confirm complex formation, measure particle size and evaluate zeta potential, as well as conduct luciferase gene reporter assays. The anti-tumor effects of pDNA ternary complexes were evaluated using suicide gene (pTK) coding thymidine kinase in Huh7-implanted mice. RESULTS: Transfection efficiencies of pDNA/chitosan/HA ternary complexes were dependent on the average molecular masses of HA. The coating of pDNA/chitosan complexes with HA maintained the cellular transfection efficiencies of lyophilized pDNA ternary complexes. Furthermore, intratumoral injection of lyophilized, rehydrated pDNA ternary complexes into tumor-bearing mice showed a significant suppression of tumor growth. CONCLUSIONS: The coating of pDNA/chitosan complexes with high-molecular-weight HA augmented the stability and cellular transfection ability of the complexes after lyophilization-rehydration.


Subject(s)
Chitosan/therapeutic use , Genetic Therapy/methods , Hyaluronic Acid/therapeutic use , Animals , DNA/genetics , Electrophoretic Mobility Shift Assay , Freeze Drying , Gene Transfer Techniques , Genes, Reporter , Genetic Vectors , Humans , Luciferases/genetics , Mice , Nanoparticles , Particle Size , Plasmids , Thymidine Kinase/genetics , Transfection
18.
Biochem Biophys Res Commun ; 483(1): 107-114, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28042030

ABSTRACT

In cancer treatment, specifically targeting cancer cells is important for optimal therapeutic efficacy. One strategy is to utilize a cancer specific promoter to express a cytotoxic gene or a viral gene required for replication. In this approach, the therapeutic window is dependent on the relative promoter activity in cancer cells versus normal cells. Therefore, a promoter with optimal cancer cell-specificity should be used. The tumor suppressor ARF promoter, which specifically responds to deregulated E2F activity, is a potent candidate. Defects in the RB pathway resulting in deregulated E2F activity are observed in almost all cancers. Furthermore, the ARF promoter exhibits greater cancer cell specificity than the E2F1 promoter and consequently, adenovirus expressing HSV-TK under the control of the ARF promoter (Ad-ARF-TK) has more selective cytotoxicity in cancer cells than the analogous E2F1 construct. Ideally, cancer specific gene expression driven by the ARF promoter could be enhanced for optimal therapeutic efficacy, with minimal side effects. We show here that ectopic expression of the CDK inhibitor p21Cip1 enhanced deregulated E2F activity and pro-apoptotic E2F target gene expression in cancer cells. Moreover, ectopic expression of p21Cip1 augmented cancer specific cytotoxicity of Ad-ARF-TK, and apoptosis induced by p21Cip1 was dependent on deregulated E2F activity. These results suggest that p21Cip1 specifically enhances deregulated E2F activity and that a combination of the CDK inhibitor with Ad-ARF-TK could be effectively employed for cancer therapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Genes, p16 , Apoptosis , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/metabolism , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Ectopic Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Promoter Regions, Genetic
19.
Cancer Cell Int ; 17: 96, 2017.
Article in English | MEDLINE | ID: mdl-29089859

ABSTRACT

BACKGROUND: Cellular based therapeutic approaches for cancer rely on careful consideration of finding the optimal cell to execute the cellular goal of cancer treatment. Cell lines and primary cell cultures have been used in some studies to compare the in vitro and in vivo efficacy of autologous vs allogeneic tumour cell vaccines. METHODS: This study examines the effect of γ-irradiation on a range of tumor cell lines in conjunction with suicide gene therapy of cancer. To determine the efficacy of this modality, a series of in vitro and in vivo experiments were conducted using genetically modified and unmodified tumor cell lines. RESULTS: Following co-culture of HSV-TK modified tumor cells and unmodified tumor cells both in vitro and in vivo we observed that the PA-STK ovarian tumor cells were sensitive to γ-irradiation, completely abolishing their ability to induce bystander killing of unmodified tumor cells. In contrast, TK-modified human and mouse mesothelioma cells were found to retain their in vitro and in vivo bystander killing effect after γ-irradiation. Morphological evidence was consistent with the death of PA-STK cells being by pyknosis after γ-irradiation. These results suggest that PA-STK cells are not suitable for clinical application of suicide gene therapy of cancer, as lethal γ-irradiation (100 Gy) interferes with their bystander killing activity. However, the human mesothelioma cell line CRL-5830-TK retained its bystander killing potential after exposure to similarly lethal γ-irradiation (100 Gy). CRL-5830 may therefore be a suitable vehicle for HSV-TK suicide gene therapy. CONCLUSIONS: This study highlights the diversity among tumor cell lines and the careful considerations needed to find the optimal tumor cell line for this type of suicide gene therapy of cancer.

20.
Mol Biol Rep ; 44(4): 323-332, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28748410

ABSTRACT

Recent reports on various cancer models demonstrate a great potential of cytosine deaminase/5-fluorocytosine suicide system in cancer therapy. However, this approach has limited success and its application to patients has not reached the desirable clinical significance. Accordingly, the improvement of this suicide system is an actively developing trend in gene therapy. The purpose of this study was to explore the cytotoxic effect observed after co-expression of hepatitis A virus 3C protease (3C) and yeast cytosine deaminase/uracil phosphoribosyltransferase fusion protein (FCU1) in a bicistronic vector. A set of mono- and bicistronic plasmid constructs was generated to provide individual or combined expression of 3C and FCU1. The constructs were introduced into HEK293 and HeLa cells, and target protein synthesis as well as the effect of 5-fluorocytosine on cell death and the time course of the cytotoxic effect was studied. The obtained vectors provide for the synthesis of target proteins in human cells. The expression of the genes in a bicistronic construct provide for the cytotoxic effect comparable to that observed after the expression of genes in monocistronic constructs. At the same time, co-expression of FCU1 and 3C recapitulated their cytotoxic effects. The combined effect of the killer and suicide genes was studied for the first time on human cells in vitro. The integration of different gene therapy systems inducing cell death (FCU1 and 3C genes) in a bicistronic construct allowed us to demonstrate that it does not interfere with the cytotoxic effect of each of them. A combination of cytotoxic genes in multicistronic vectors can be used to develop pluripotent gene therapy agents.


Subject(s)
Cysteine Endopeptidases/biosynthesis , Cytosine Deaminase/biosynthesis , Flucytosine/pharmacology , Genetic Therapy/methods , Hepatitis A Virus, Human/enzymology , Pentosyltransferases/biosynthesis , Viral Proteins/biosynthesis , 3C Viral Proteases , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Flucytosine/pharmacokinetics , Genes, Transgenic, Suicide , Genetic Vectors , HEK293 Cells , HeLa Cells , Hepatitis A Virus, Human/metabolism , Humans , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Plasmids/genetics , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Transduction, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL