1
|
Szewczyk-Roszczenko O, Barlev NA. The Role of p53 in Nanoparticle-Based Therapy for Cancer. Cells 2023; 12:2803. [PMID: 38132123 PMCID: PMC10742014 DOI: 10.3390/cells12242803] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
p53 is arguably one of the most important tumor suppressor genes in humans. Due to the paramount importance of p53 in the onset of cell cycle arrest and apoptosis, the p53 gene is found either silenced or mutated in the vast majority of cancers. Furthermore, activated wild-type p53 exhibits a strong bystander effect, thereby activating apoptosis in surrounding cells without being physically present there. For these reasons, p53-targeted therapy that is designed to restore the function of wild-type p53 in cancer cells seems to be a very appealing therapeutic approach. Systemic delivery of p53-coding DNA or RNA using nanoparticles proved to be feasible both in vitro and in vivo. In fact, one p53-based therapeutic (gendicine) is currently approved for commercial use in China. However, the broad use of p53-based therapy in p53-inactivated cancers is severely restricted by its inadequate efficacy. This review highlights the current state-of-the-art in this area of biomedical research and also discusses novel approaches that may help overcome the shortcomings of p53-targeting nanomedicine.
Collapse
Affiliation(s)
- Olga Szewczyk-Roszczenko
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Nikolai A. Barlev
- Department of Biomedicine, School of Medicine, Nazarbayev University, Kerey and Zhanibek Khans St., Astana 020000, Kazakhstan
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow 119121, Russia
- Institute of Cytology, 4 Tikhoretsky Ave., Saint-Petersburg 194064, Russia
| |
Collapse
|
2
|
Xu S, He J, Imtiyaz Z, Agrawal AK, Woodle MC, Mixson AJ. Marked increase in tumor transfection with a truncated branched polymer. J Gene Med 2021; 24:e3396. [PMID: 34713552 PMCID: PMC8724455 DOI: 10.1002/jgm.3396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/08/2021] [Accepted: 10/16/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND We previously determined that polyplexes formed by linear H2K peptides were more effective in transfecting tumors in vivo than polyplexes formed by branched H2K4b-20 peptides. Based on trypsin digest and salt displacement studies, the linear H2K polyplexes were less stable than the branched H2K4b-20 polyplexes. Because binding and release of the polymer and DNA from the H2K4b-20 polyplex may account for the ineffectiveness, we investigated whether four-branched histidine-lysine (HK) peptides with varying numbers of amino acids in their branches would be more effective in their ability to increase gene expression in tumors in vivo. METHODS Linear and branched peptides with multiple -KHHK- motifs were synthesized by solid-phase synthesis. The branched H2K4b-20, -18, -14 and 12 peptides had 20, 18, 14 and 12 amino acids in their branches, respectively. These peptides were examined for their ability to carry luciferase-expressing plasmids to human breast cancer xenografts in a mouse model. With gel retardation and in vivo transfection, the incorporation of a targeting ligand and an endosomal lysis peptide into these polyplexes was also examined. A blocking antibody was pre-injected prior to the polyplexes to determine the role of neuropilin 1 in the uptake of these polyplexes by the tumor. The size of the polyplexes was measured by dynamic light scattering. RESULTS Of the four negative surface-charge polyplexes formed by the branched carriers, the H2K4b-14 polyplex was determined to be the most effective plasmid delivery platform to tumors. The incorporation of a targeting ligand and an endosomal lysis peptide into H2K4b-14 polyplexes further enhanced their ability to transfect tumors in vivo. Furthermore, after pre-injecting tumor-bearing mice with a blocking antibody to the neuropilin-1 receptor (NRP-1), there was a marked reduction of tumor gene expression with the modified H2K4b-14 polyplexes, suggesting that NRP-1 mediated their transport into the tumor. CONCLUSIONS The present study established that branched peptides intermediate in length were very efficient in delivering plasmids to tumors in vivo.
Collapse
Affiliation(s)
- Songhui Xu
- Department of Pathology, University Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Jiaxi He
- Department of Pathology, University Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Zuha Imtiyaz
- Department of Pathology, University Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Atul K Agrawal
- Department of Pathology, University Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| | | | - A James Mixson
- Department of Pathology, University Maryland School of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
3
|
HEGER ZBYNEK, GUMULEC JAROMIR, CERNEI NATALIA, TMEJOVA KATERINA, KOPEL PAVEL, BALVAN JAN, MASARIK MICHAL, ZITKA ONDREJ, BEKLOVA MIROSLAVA, ADAM VOJTECH, KIZEK RENE. 17β-estradiol-containing liposomes as a novel delivery system for the antisense therapy of ER-positive breast cancer: An in vitro study on the MCF-7 cell line. Oncol Rep 2014; 33:921-9. [DOI: 10.3892/or.2014.3627] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/11/2014] [Indexed: 11/06/2022] Open
|
4
|
Targeting tumors using nanoparticle platforms: a phase I study of a systemically administered gene therapy system. Mol Ther 2014; 21:922-3. [PMID: 23636291 DOI: 10.1038/mt.2013.76] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
5
|
Abstract
Induction of cytokines by small interfering RNA (siRNA) polyplexes has been a significant concern of researchers attempting to minimize the toxicity of this promising therapy. Although cationic carriers of siRNA are known to increase cytokine levels, few systematic studies have been done to determine what properties of the carrier are important to modulate cytokines. Because branched histidine-lysine (HK) peptides are effective carriers of siRNA and their sequence can be readily modified, we selected this class of carrier to determine which sequences of the peptide were important for cytokine induction. With the use of peripheral blood mononuclear cells (PBMCs), the HK peptide with a higher number of histidines (H3K(+H)4b) in complex with siRNA induced lower levels of cytokines compared with other HK (e.g., H2K4b, H3K4b, H3K(+N)4b) siRNA nanoplexes. Notably, these peptides' siRNA polyplexes showed a similar pattern of cytokine induction when injected intravenously in a mouse model, i.e., the HK with higher content of histidines induced cytokines the least. As indicated by the pH-sensitive dye within acidic endosomes, the greater pH-buffering capacity of H3K(+H)4b compared with other HK peptides may explain why cytokine levels were reduced. In addition to buffering capacity, the size of HK polyplexes markedly influenced cytokine production.
Collapse
|
6
|
Prabha S, Sharma B, Labhasetwar V. Inhibition of tumor angiogenesis and growth by nanoparticle-mediated p53 gene therapy in mice. Cancer Gene Ther 2012; 19:530-7. [PMID: 22595792 PMCID: PMC3400709 DOI: 10.1038/cgt.2012.26] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutation of the p53 tumor suppressor gene, the most common genetic alteration in human cancers, results in more aggressive disease and increased resistance to conventional therapies. Aggressiveness may be related to the increased angiogenic activity of cancer cells containing mutant p53. To restore wild-type p53 function in cancer cells, we developed polymeric nanoparticles (NPs) for p53 gene delivery. Previous in vitro and in vivo studies demonstrated the ability of these NPs to provide sustained intracellular release of DNA, thus sustained gene transfection and decreased tumor cell proliferation. We investigated in vivo mechanisms involved in NP-mediated p53 tumor inhibition, with focus on angiogenesis. We hypothesize that sustained p53 gene delivery will help decrease tumor angiogenic activity and thus reduce tumor growth and improve animal survival. Xenografts of p53 mutant tumors were treated with a single intratumoral injection of p53NPs. We observed intratumoral p53 gene expression corresponding to tumor growth inhibition, over 5 weeks. Treated tumors showed upregulation of thrombospondin-1, a potent antiangiogenic factor, and a decrease in microvessel density vs. controls (saline, p53 DNA alone, and control NPs). Greater levels of apoptosis were also observed in p53NP-treated tumors. Overall, this led to significantly improved survival in p53NP-treated animals. NP-mediated p53 gene delivery slowed cancer progression and improved survival in an in vivo cancer model. One mechanism by which this is accomplished is disruption of tumor angiogenesis. We conclude that the NP-mediated sustained tumor p53 gene therapy can effectively be used for tumor growth inhibition.
Collapse
Affiliation(s)
- S Prabha
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, USA
| | | | | |
Collapse
|
7
|
Abstract
Genome editing with the use of zinc finger nucleases has been successfully applied to variety of a eukaryotic cells. Furthermore, the proof of concept for this approach has been extended to diverse animal models from Drosophila to mice. Engineered zinc finger nucleases are able to target specifically and manipulate disease-causing genes through site-specific double strand DNA breaks followed by non-homologous end joining or homologous recombination mechanisms. Consequently, this technology has considerable flexibility that can result in either a gain or loss of function of the targeted gene. In addition to this flexibility, gene therapy by zinc finger nucleases may enable persistent long term gene modification without continuous transfection- a potential advantage over RNA interference or direct gene inhibitors. With systemic viral delivery systems, this gene-editing approach corrected the mutant factor IX in models of mouse hemophilia. Moreover, phase I clinical trials have been initiated with zinc finger nucleases in patients with glioblastoma and HIV. Thus, this emerging field has significant promise as a therapeutic strategy for human genetic diseases, infectious diseases and oncology. In this article, we will review recent advances and potential risks in zinc finger nuclease gene therapy.
Collapse
Affiliation(s)
- S-T Chou
- Department of Pathology, University of Maryland School of Medicine, MSTF Building, 10 South Pine Street, Baltimore, MD 21201, USA ; Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742
| | | | | |
Collapse
|
8
|
Sasaki Y, Negishi H, Idogawa M, Yokota I, Koyama R, Kusano M, Suzuki H, Fujita M, Maruyama R, Toyota M, Saito T, Tokino T. p53 negatively regulates the hepatoma growth factor HDGF. Cancer Res 2011; 71:7038-47. [PMID: 22006999 DOI: 10.1158/0008-5472.can-11-1053] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hepatoma-derived growth factor (HDGF) is a secreted heparin-binding growth factor that has been implicated in cancer development and progression. Here, we report that HDGF is a critical target for transcriptional repression by the tumor suppressor p53. Endogenous HDGF expression was decreased in cancer cells with introduction of wild-type p53, which also downregulated HDGF expression after DNA damage. In support of the likelihood that HDGF is a critical driver of cancer cell growth, addition of neutralizing HDGF antibodies to culture media was sufficient to block cell growth, migration, and invasion. Similarly, these effects were elicited by conditioned culture medium from p53-expressing cells, and they could be reversed by the addition of recombinant human HDGF. Interestingly, we found that HDGF was overexpressed also in primary gastric, breast, and lung cancer tissues harboring mutant p53 genes. Mechanistic investigations revealed that p53 repressed HDGF transcription by altering HDAC-dependent chromatin remodeling. Taken together, our results reveal a new pathway in which loss of p53 function contributes to the aggressive pathobiological potential of human cancers by elevating HDGF expression.
Collapse
Affiliation(s)
- Yasushi Sasaki
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Sharma B, Ma W, Adjei IM, Panyam J, Dimitrijevic S, Labhasetwar V. Nanoparticle-mediated p53 gene therapy for tumor inhibition. Drug Deliv Transl Res 2011; 1:43-52. [PMID: 22553503 PMCID: PMC3339849 DOI: 10.1007/s13346-010-0008-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The p53 tumor suppressor gene is mutated in 50% of human cancers, resulting in more aggressive disease with greater resistance to chemotherapy and radiation therapy. Advances in gene therapy technologies offer a promising approach to restoring p53 function. We have developed polymeric nanoparticles (NPs), based on poly (lactic-co-glycolic acid), that provide sustained intracellular delivery of plasmid DNA, resulting in sustained gene expression without vector-associated toxicity. Our previous studies with p53 gene-loaded NPs (p53NPs) demonstrated sustained antiproliferative effects in cancer cells in vitro. The objective of this study was to evaluate the efficacy of p53NPs in vivo. Tumor xenografts in mice were established with human p53-null prostate cancer cells. Animals were treated with p53NPs by either local (intratumoral injection) or systemic (intravenous) administration. Controls included saline, p53 DNA alone, and control NPs. Mice treated with local injections of p53NPs demonstrated significant tumor inhibition and improved animal survival compared with controls. Tumor inhibition corresponded to sustained and greater p53 gene and protein expression in tumors treated with p53NPs than with p53 DNA alone. A single intravenous dose of p53NPs was successful in reducing tumor growth and improving animal survival, although not to the same extent as with local injections. Imaging studies showed that NPs accumulate in tumor tissue after intravenous injection; however, further improvement in tumor targeting efficiency of p53NPs may be needed for better outcome. In conclusion, the NP-mediated p53 gene therapy is effective in tumor growth inhibition. NPs may be developed as nonviral vectors for cancer and other genetic diseases.
Collapse
Affiliation(s)
- Blanka Sharma
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Wenxue Ma
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Isaac Morris Adjei
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Jayanth Panyam
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Sanja Dimitrijevic
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Vinod Labhasetwar
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
10
|
Xiao W, Chen X, Yang L, Mao Y, Wei Y, Chen L. Co-delivery of doxorubicin and plasmid by a novel FGFR-mediated cationic liposome. Int J Pharm 2010; 393:119-26. [PMID: 20416367 DOI: 10.1016/j.ijpharm.2010.04.018] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 04/14/2010] [Accepted: 04/15/2010] [Indexed: 02/05/2023]
Abstract
In our previous study, we developed a novel cationic liposome, which was modified with truncated human basic fibroblast growth factor (tbFGF) peptide. This tbFGF-mediated cationic liposome could deliver chemotherapeutic agents or gene specifically to FGFRs on tumors and obtained higher transfection efficiency than plain cationic liposomes. In order to investigate whether this novel cationic liposome could achieve a synergistic/combined anti-tumor effect as a co-delivery system, we simultaneously delivered doxorubicin (DOX) and the plasmid encoding the phosphorylation-defective mouse survivin threonine 34-->alanine mutant (Msurvivin T34A plasmid) to the same cells through this cationic liposome. As a result, an enhanced antiproliferative activity in vitro has been achieved by delivering DOX and DNA simultaneously to the Lewis lung carcinoma cells (LLC) using this liposome. The concentration of DOX in the co-delivery system which caused 50% killing was nearly 3-fold lower than that of the free DOX. Furthermore, the co-delivery system suppressed tumor growth more efficiently than either DOX or the Msurvivin T34A plasmid alone in the Lewis lung carcinoma-bearing C57BL/6 mice. After 18 days of treatment with the co-delivery system, the average tumor volume in mice was decreased by 80%, which was higher than liposomal DOX (70%, P<0.05) and Msurvivin T34A plasmid (41%, P<0.01). The co-delivery system also caused 15 days delay of tumor growth, which was longer than the other treatment groups. In conclusion, this novel cationic liposome is an efficient vector to simultaneously deliver drugs and DNA to the same cells in vitro and in vivo.
Collapse
Affiliation(s)
- Wenjing Xiao
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | |
Collapse
|
11
|
You Y, Xue X, Li M, Qin X, Zhang C, Wang W, Giang C, Wu S, Liu Y, Zhu W, Ran Y, Zhang Z, Han W, Zhang Y. Inhibition effect of pcDNA-tum-5 on the growth of S180 tumor. Cytotechnology 2007; 56:97-104. [PMID: 19002847 DOI: 10.1007/s10616-007-9117-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Accepted: 12/18/2007] [Indexed: 10/22/2022] Open
Abstract
Tumor growth and metastasis depend on vessel formation, and inhibition of angiogenesis of tumor by production of anti-angiogenic drugs should be a promising approach for cancer therapy. Tumstatin is an angiogenesis inhibitor. The anti-angiogenic activity of tumstatin is localized to the 54-132 amino acids. The gene fragment encoding amino acids 45-132 of tumstatin (tum-5) was subcloned into pcDNA3.1 (pcDNA-tum5). Tum-5 protein could be expressed and secreted in CHO cells after transfection. The conditioned medium (containing tum-5 protein) from the transfectant has an anti-angiogenic effect on HUVEC cells in vitro. The anti-tumor effect of pcDNA-tum5 on mice bearing S180 tumors was evaluated. The results showed that pcDNA-tum-5 has a significant inhibition activity in the growth of the tumors. This study suggests that the gene delivery of tum-5 may be an effective strategy for cancer therapy.
Collapse
Affiliation(s)
- Yanjie You
- Biotechnology Center of The Fourth Military Medical University, 17 Changle West Road, 710032, Xi'an, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Chhipa RR, Bhat MK. Bystander killing of breast cancer MCF-7 cells by MDA-MB-231 cells exposed to 5-fluorouracil is mediated via Fas. J Cell Biochem 2007; 101:68-79. [PMID: 17340621 DOI: 10.1002/jcb.21153] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The major drawback with cancer therapy is the development of resistant cells within tumors due to their heterogeneous nature and due to inadequate drug delivery during chemotherapy. Therefore, the propagation of injury ("bystander effect" (BE)) from directly damaged cells to other cells may have great implications in cancer chemotherapy. The general advantage of the bystander cell killing phenomenon is the large therapeutic index that can be achieved. Experiments suggest that this phenomenon is detected in radiation therapy as well as in gene therapy in conjunction with chemotherapy. In the present study, we developed an original in vitro model dedicated to the exploration of bystander cytotoxicity induced during breast carcinoma chemotherapy. In brief, we investigated this perpetuation of injury on untreated bystander MCF-7 breast cancer cells which were coplated with 5-fluorouracil (5-FU)-treated MDA-MB-231 breast cancer cells. To achieve this goal, a specific in vitro coculture model which involved mixing of aggressive MDA-MB-231 breast cancer cells with enhanced green fluorescent protein (EGFP) expressing stable clone of non-metastatic MCF-7 breast cancer cells (MCF-EGFP), was used. A bystander killing effect was observed in MCF-EGFP cells cocultured with MDA-MB-231 cells pretreated with 5-FU. The striking decrease in MCF-EGFP cells, as detected by assaying for total GFP intensity, is mediated by activation of Fas/FasL system. The implication of Fas in MCF-EGFP cell death was confirmed by using antagonistic anti-FasL antibody that reverses bystander cell death by blocking FasL on MDA-MB-231 cells. In addition, inhibition of CD95/Fas receptor on the cell surface of MCF-EGFP cells by treatment with Pifithrin-alpha, a p53 specific transactivation inhibitor, partially abrogated the sensitivity of bystander MCF-EGFP cells. Our data, therefore, demonstrates that the Fas/FasL system could be considered as a new determinant for chemotherapy-induced bystander cell death in breast cancers.
Collapse
Affiliation(s)
- Rishi Raj Chhipa
- National Centre for Cell Science, Ganeshkhind, Pune 411 007, India
| | | |
Collapse
|
13
|
Collinet P, Vereecque R, Sabban F, Vinatier D, Leblanc E, Narducci F, Querleu D, Quesnel B. In vivo expression and antitumor activity of p53 gene transfer with naked plasmid DNA in an ovarian cancer xenograft model in nude mice. J Obstet Gynaecol Res 2006; 32:449-53. [PMID: 16984510 DOI: 10.1111/j.1447-0756.2006.00435.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Abnormalities in the p53 and p16 tumor suppressor genes are one of the most common occurrences associated with human neoplasia. Consequently, restoration of wild-type p53 or p16 functions is seen as a particularly promising approach for cancer gene therapy. In vitro and in vivo data have demonstrated that virus-mediated p53 gene transfer can induce active cell death and ovarian tumor regression. AIM To evaluate the efficiency of intratumoral injection of naked DNA in tumor growth inhibition in an ovarian xenograft model. For that purpose, plasmid vectors encoding wild-type p53 (wt-p53) or p16 alone or in combination were used. METHODS Nude mice were injected subcutaneously with the human ovarian adenocarcinoma cell line SKOV3. Three weeks after xenograft, tumor-bearing mice were injected twice a week with plasmid vectors carrying WT-p53 and/or WT-p16 cDNA. Empty plasmids and saline buffer were used as control. Tumor growth was monitored to evaluate the inhibition potential with p53 and/or p16 restoration. RESULTS When compared to the control, intratumoral repeated injections of naked plasmid DNA encoding wt-p53 were inhibiting tumor growth. This inhibition was not observed with p16 and no synergy could be obtained between p53 and p16. p53 expression was restored in 84% of mice injected with plasmid encoding wt-p53. p16 expression was restored in 63% of mice injected with plasmid encoding p16. CONCLUSIONS In this report we demonstrated that: (i) naked DNA represents an efficient gene transfer in the SKOV3 xenograft model; (ii) restoration of wt-p53 gene allows tumor growth inhibition; and (iii) this inhibition could be correlated with p53 expression as seen in 84% of treated mice after repeated naked DNA injections. These results allow us to envisage naked DNA as a therapeutic adjuvant in ovarian cancer treatment, concomitantly with tumor resection and chemotherapy.
Collapse
Affiliation(s)
- Pierre Collinet
- Hôpital Jeanne de Flandre, Clinique de Gynécologie-Obstétrique CHRU Lille, Lille Cedex, France.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
To date, dysfunctional tumour suppressor genes are the most common genetic lesions identified in human cancers. Functional copies of tumour suppressor genes can be introduced into cancer cells by gene transfer using adenoviral vectors. This approach has been extensively studied in the clinic with intratumoural injection of a replication-defective adenovirus that expresses p53 (Ad-p53). Overexpression of p53 in cancer cells induces growth arrest and apoptosis. Ad-p53 injections have an excellent safety profile, and have mediated tumour regression and growth arrest as monotherapy, or have overcome resistance or increased the effectiveness of radiation therapy and chemotherapy. Expression of the p53 transgene has occurred at high levels and is associated with the activation of other genes in the p53 pathway. These studies indicate proof-of-principle for tumour suppressor gene therapy and represent a new paradigm in targeted therapy.
Collapse
Affiliation(s)
- Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, UT M. D. Anderson Cancer Center, P.O. Box 301402, Houston, TX 77230-1402, USA.
| |
Collapse
|
15
|
Abstract
In view of the limited success of available treatment modalities for breast cancer, alternative and complementary strategies need to be developed. The delineation of the molecular basis of breast cancer provides the possibility of specific intervention by gene therapy through the introduction of genetic material for therapeutic purposes. In this regard, several gene therapy approaches for carcinoma of the breast have been developed. These approaches can be divided into six broad categories: (1) mutation compensation, (2) molecular chemotherapy, (3) proapoptotic gene therapy, (4) antiangiogenic gene therapy, (5) genetic immunopotentiation, and (6) genetic modulation of resistance/sensitivity. Clinical trials for breast cancer have been initiated to evaluate safety, toxicity, and efficacy. Combined modality therapy with gene therapy and chemotherapy or radiation therapy has shown promising results. It is expected that as new therapeutic targets and approaches are identified and advances in vector design are realized, gene therapy will play an increasing role in clinical breast cancer treatment.
Collapse
Affiliation(s)
- MA Stoff-Khalili
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birminham, AL, USA
- Department of Obstetrics and Gynecology, University of Duesseldorf, Medical Center, Duesseldorf, Germany
| | - P Dall
- Department of Obstetrics and Gynecology, University of Duesseldorf, Medical Center, Duesseldorf, Germany
| | - DT Curiel
- Division of Human Gene Therapy, Departments of Medicine, Surgery, Pathology and the Gene Therapy Center, University of Alabama at Birmingham, Birminham, AL, USA
| |
Collapse
|
16
|
Leng Q, Scaria P, Ioffe OB, Woodle M, Mixson AJ. A branched histidine/lysine peptide, H2K4b, in complex with plasmids encoding antitumor proteins inhibits tumor xenografts. J Gene Med 2006; 8:1407-15. [PMID: 17133339 DOI: 10.1002/jgm.982] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND In this study we investigated whether a particular branched HK polymer, H2K4b, was an effective in vivo carrier of plasmids expressing the antiangiogenic kringle 1-5 or the tumor suppressor p53. METHODS H2K4b was synthesized on a solid-phase peptide synthesizer. Distribution, optimization and time course studies were done in tumor-bearing nude mice by systemically administering H2K4b in complex with a luciferase-expressing plasmid. We examined the amount of tumor angiogenesis in C6 with MDA-MB-435 xenografts utilizing the carmine dye. The ability of H2K4b to carry luciferase plasmids to different tissues was compared with several liposomal carriers. Medium from cells transfected with mKr1-5 was tested for its capacity to inhibit angiogenesis with an in vivo Matrigel assay. We then determined if systemically delivered H2K4b in complex with plasmid encoding mKr1-5 inhibited tumor growth; we also compared the antitumor activity of HK polyplexes containing hKr1-5, mKr1-5, and p53 plasmids. RESULTS H2K4b carried the luciferase-expressing plasmid in order of descending efficacy to these tissues: lung, spleen, tumor, and liver. Compared to DOTAP-containing liposomes, H2K4b was a more effective carrier of a luciferase-containing plasmid to extrapulmonary tissues. We then determined that mKr1-5 in complex with H2K4b reduced MDA-MB-435 tumor growth by approximately 50% compared to the control group (P < 0.01). Similarly, H2K4b/mKr1-5 polyplexes reduced the growth of C6 xenografts. In MDA-MB-435 xenografts, p53- and Kr1-5-expressing plasmids in complex with H2K4b had comparable antitumor activity. CONCLUSION H2K4b demonstrates potential as a carrier of plasmids encoding antiangiogenic and/or tumor suppressor proteins in a tumor-bearing mouse model.
Collapse
Affiliation(s)
- Qixin Leng
- Department of Pathology, University of Maryland Baltimore, MSTF Building, 10 South Pine Street, Baltimore, MD 21201, USA
| | | | | | | | | |
Collapse
|
17
|
Kang X, Xiao X, Harata M, Bai Y, Nakazaki Y, Soda Y, Kurita R, Tanaka T, Komine F, Izawa K, Kunisaki R, Setoyama M, Nishimori H, Natsume A, Sunamura M, Lozonshi L, Saitoh I, Tokino T, Asano S, Nakamura Y, Tani K. Antiangiogenic activity of BAI1 in vivo: implications for gene therapy of human glioblastomas. Cancer Gene Ther 2005; 13:385-92. [PMID: 16244591 DOI: 10.1038/sj.cgt.7700898] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glioblastomas are the most common primary brain tumors in adults. These tumors exhibit a high degree of vascularization, and malignant progression from astrocytoma to glioblastoma is often accompanied by increased angiogenesis and the upregulation of vascular endothelial growth factor and its receptors. In this study, we investigated the in vivo antiangiogenic and antitumor effects of brain-specific angiogenesis inhibitor 1 (BAI1) using human glioblastoma cell lines. Glioblastoma cells were transduced with an adenoviral vector encoding BAI1 (AdBAI1), and Northern and Western blot analyses, respectively, demonstrated BAI1 mRNA and protein expression in the transduced tumor cells. Using an in vivo neovascularization assay, we found that angiogenesis surrounding AdBAI1-transduced glioblastoma cells transplanted into transparent skinfold chambers of SCID mice was significantly impaired compared to control treated cells. Additionally, in vivo inoculation with AdBAI1 of established subcutaneous or intracerebral transplanted tumors significantly impaired tumor growth and promoted increased mouse survival. Morphologically, the tumors exhibited signs of impaired angiogenesis, such as extensive necrosis and reduced intratumoral vascular density. Taken together, these data strongly indicate that BAI1 may be an excellent gene therapy candidate for the treatment of brain tumors, especially human glioblastomas.
Collapse
Affiliation(s)
- X Kang
- Division of Molecular Therapy, Advanced Clinical Research Center, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Dickson PV, Nathwani AC, Davidoff AM. Delivery of antiangiogenic agents for cancer gene therapy. Technol Cancer Res Treat 2005; 4:331-41. [PMID: 16029054 DOI: 10.1177/153303460500400403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The understanding that tumor growth and metastasis are angiogenesis dependent processes has led to interest in targeting tumor vasculature in anticancer therapy. Furthermore, recent insights into the molecular interactions that orchestrate physiologic and pathologic angiogenesis have resulted in a variety of antiangiogenic strategies. A gene therapy-mediated approach for the delivery of antiangiogenic agents has several advantages, including the potential for sustained expression. However, the choice of angiogenesis inhibitor, method of gene delivery, and target/site for transgene expression are important variables to be considered when designing this approach. Here we review the major alternatives within each of these categories and provide illustrative examples of their use in preclinical models.
Collapse
Affiliation(s)
- Paxton V Dickson
- Department of Surgery, St. Jude Children's Research Hospital, and the Department of Surgery, The University of Tennessee-Memphis, Health Science Center, TN 38163, USA
| | | | | |
Collapse
|
19
|
Nakase M, Inui M, Okumura K, Kamei T, Nakamura S, Tagawa T. p53 gene therapy of human osteosarcoma using a transferrin-modified cationic liposome. Mol Cancer Ther 2005; 4:625-31. [PMID: 15827336 DOI: 10.1158/1535-7163.mct-04-0196] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gene delivery via transferrin receptors, which are highly expressed by cancer cells, can be used to enhance the effectiveness of gene therapy for cancer. In this study, we examined the efficacy of p53 gene therapy in human osteosarcoma (HOSM-1) cells derived from the oral cavity using a cationic liposome supplemented with transferrin. HOSM-1 cells were exposed to transferrin-liposome-p53 in vitro, and the growth inhibition rate, expression of p53 and bax, and induction of apoptosis were measured 48 hours later. Treatment of HOSM-1 cells with transferrin-liposome-p53 resulted in 60.7% growth inhibition. Wild-type p53 expression and an increase in bax expression were observed following transfection with transferrin-liposome-p53, and 20.5% of the treated HOSM-1 cells were apoptotic. In vivo, the HOSM-1 tumor transplanted into nude mice grew to 5 to 6 mm in diameter. Following growth of the tumor to this size, transferrin-liposome-p53 was locally applied to the peripheral tumor (day 0) and then applied once every 5 days for a total of six times. During the administration period, tumor growth did not occur, and the mean tumor volume on the last day of administration (day 25) was 10.0% of that in the saline control group. These results suggest that p53 gene therapy via cationic liposome modification with transferrin is an effective strategy for treatment of osteosarcoma.
Collapse
Affiliation(s)
- Minoru Nakase
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Mie University, 2-174 Edobashi, Tsu 514-8507, Japan.
| | | | | | | | | | | |
Collapse
|
20
|
El-Aneed A. Current strategies in cancer gene therapy. Eur J Pharmacol 2005; 498:1-8. [PMID: 15363969 DOI: 10.1016/j.ejphar.2004.06.054] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2004] [Accepted: 06/29/2004] [Indexed: 10/26/2022]
Abstract
Cancer gene therapy is the most studied application of gene therapy. Many genetic alterations are involved in the transformation of a normal cell into a neoplastic one. The two main gene groups involved in cancer development are oncogenes and tumor suppressor genes. While the latter eliminates cancerous cells via apoptosis, the former enhances cell proliferation. Therefore, apoptotic genes and anti-oncogenes are widely used in cancer gene therapy. In addition to oncogenes and tumor suppressor genes, chemotherapy and gene therapy can be combined through suicide gene strategy. A suicide gene encodes for a non-mammalian enzyme; this enzyme is used to convert a non-toxic prodrug into its active cytotoxic metabolite within the cancerous cells. Tumor suppressor genes, anti-oncogenes and suicide genes target cancer cells on the molecular level. On the other hand, cancer is immunogenic in nature; therefore, it can also be targeted on the immunological level. Boosting the immune response against cancerous cells is usually achieved via genes encoding for cytokines. Interleukin-12 gene, for example, is one of the most studied cytokine genes for cancer gene therapy applications. DNA vaccines are also used after conventional treatments to eliminate remnant malignant cells. All these therapeutic strategies and other strategies namely anti-angiogenesis and drug resistant genes are briefly reviewed and highlighted in this article.
Collapse
Affiliation(s)
- Anas El-Aneed
- Biochemistry Department, Memorial University of Newfoundland, St. John's, NL, Canada A1B 3X9.
| |
Collapse
|
21
|
Leng Q, Mixson AJ. Modified branched peptides with a histidine-rich tail enhance in vitro gene transfection. Nucleic Acids Res 2005; 33:e40. [PMID: 15731333 PMCID: PMC549579 DOI: 10.1093/nar/gni040] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Successful gene therapy depends on the development of efficient, non-toxic gene delivery systems. To accomplish this objective, our laboratory has focused on solid-phase synthesized peptide carriers, in which the amino acid sequence can be varied precisely to augment intracellular DNA transport. We previously determined that linear and branched co-polymers of histidine and lysine in combination with liposomes enhanced the efficiency of gene transfection. In this study, we have modified two branched histidine-lysine (HK) peptides by adding a histidine-rich tail. In a variety of cell lines, this histidine-rich tail markedly improved transfection efficiency, presumably by increasing the buffering capacity of the polymer. One polymer with a histidine-rich tail, H2K4bT, compared favorably with the commonly used transfection agents. Together with modification of our transfection protocol, these improved HK peptides alone, without liposomes, are the effective carriers of plasmids into a variety of cells. We anticipate that branched HK peptides will continue to be developed as carriers of nucleic acids for in vitro and in vivo applications.
Collapse
Affiliation(s)
| | - A. James Mixson
- To whom correspondence should be addressed. Tel: +1 410 706 3223; Fax: +1 410 706 8414;
| |
Collapse
|
22
|
Abstract
Current therapy such as radiation and chemotherapy controls less than 50% of lung cancers, summoning the development of novel therapeutic strategies that can directly target the underlying mechanisms of tumorigenesis. The clinical trials summarized in this article clearly demonstrate that contrary to initial predictions that gene therapy would not be suitable for cancer, gene replacement therapy is a viable potential addition to the arsenal for cancer. Gene expression has been documented and occurs even in the presence of an antiadenovirus immune response. Clinical trials have demonstrated that direct intratumor injection can cause tumor regression or prolonged stabilization of local disease, and the low toxicity associated with gene transfer indicates that tumor suppressor gene replacement can be readily combined with existing and future treatments. Initial concerns that the wide diversity of genetic lesions in cancer cells would prevent the application of gene therapy to cancer appear unfounded; on the contrary, correction of a single genetic lesion has resulted in significant tumor regression. Studies combining transfer of tumor suppressor genes with conventional DNA-damaging treatments indicate that correction of a defect in apoptosis induction can restore sensitivity to radiation and chemotherapy in some resistant tumors, and indications that sensitivity to killing might be enhanced in already sensitive tumors may eventually lead to reduced toxicity from chemotherapy and radiation therapy. The most recent data from the laboratory demonstrating damage to tumor suppressor genes in normal tissue and premalignant lesions even suggest that these genes may someday be useful in early intervention, diagnosis, and even prevention of cancer. Despite the obvious promise evident in the results of these studies, however, it is critical to recognize that there are still gaps in knowledge and technology to address. At the current rate of biotechnology development, it is only a matter of time until technical limitations that currently prevent the widespread application of gene therapy to cancer are overcome by development of more efficient vectors, discovery of novel genes, and development of combined modality approaches.
Collapse
Affiliation(s)
- Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas, M.D. Anderson Cancer Center, Box 445, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | | |
Collapse
|
23
|
Nemunaitis J, Nemunaitis J. Granulocyte-macrophage colony-stimulating factor gene-transfected autologous tumor cell vaccine: focus[correction to fcous] on non-small-cell lung cancer. Clin Lung Cancer 2004; 5:148-57. [PMID: 14667270 DOI: 10.3816/clc.2003.n.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Traditionally, non-small-cell lung cancer (NSCLC) is not thought of as an immunosensitive malignancy. However, recent clinical results with GVAX, a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transduced autologous tumor vaccine, may suggest otherwise. This review summarizes immune-induced activity caused by GM-CSF protein and GM-CSF gene-transfected vaccines. Initial indication of use for GM-CSF protein (sargramostim) was to improve neutrophil recovery following cytotoxic chemotherapy. However, several trials involving patients with hematologic malignancy demonstrated improvement in survival related to delayed disease progression in patients receiving sargramostim in combination with chemotherapy. Subsequently, others explored potential antitumor activity with sargramostim in a variety of trials. Results did not consistently demonstrate sufficient antitumor activity to justify routine use of sargramostim as an anticancer agent. Preclinical work with GM-CSF gene-transfected vaccines, however, did demonstrate significant activity, thereby justifying clinical investigation. Patients with metastatic NSCLC who had previously failed chemotherapy demonstrated response to GVAX (3 of 33 complete responses) and dose-related improvement in survival (471 days vs. 174 days).
Collapse
|
24
|
Abstract
Viruses capable of inducing lysis of malignant cells through their replication process are known as "oncolytic" viruses. Clinical trials in oncology have been performed with oncolytic viruses for nearly fifty years. Both systemic and intratumoral routes of administration have been explored. Toxicity has generally been limited to injection site pain, transient fever, and tumor necrosis. Responses with early crude materials were usually short in duration; however, recent trials with gene-attenuated viruses suggest a more prolonged duration of responses.
Collapse
|
25
|
Mhashilkar A, Chada S, Roth JA, Ramesh R. Gene therapy. Therapeutic approaches and implications. Biotechnol Adv 2004; 19:279-97. [PMID: 14538077 DOI: 10.1016/s0734-9750(01)00063-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The present article is an overview of gene therapy with an emphasis on different approaches and its implications in the clinic. Genetic interventions have been applied to the diagnosis of and therapy for an array of human diseases. The initial concept of gene therapy was focused on the treatment of genetic diseases. Subsequently, the field of gene therapy has been expanded, with a major focus on cancer. Although the results of early gene therapy-based clinical trials have been encouraging, there is a need for gene delivery vectors that feature reduced immunogenicity and improved targeting ability. The results of phases I/II clinical trials have suggested the important role of gene therapy as a versatile and powerful treatment tool, especially for human cancers. One reasonable expectation is that performing gene therapy at an earlier stage in the disease process or for minimal residual disease may be more advantageous.
Collapse
Affiliation(s)
- A Mhashilkar
- Introgen Therapeutics, Inc., Houston, TX 77030, USA
| | | | | | | |
Collapse
|
26
|
Xiang JJ, Tang JQ, Zhu SG, Nie XM, Lu HB, Shen SR, Li XL, Tang K, Zhou M, Li GY. IONP-PLL: a novel non-viral vector for efficient gene delivery. J Gene Med 2003; 5:803-17. [PMID: 12950071 DOI: 10.1002/jgm.419] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Non-viral methods of gene delivery have been an attractive alternative to virus-based gene therapy. However, the vectors that are currently available have drawbacks limiting their therapeutic application. METHODS We have developed a self-assembled non-viral gene carrier, poly-L-lysine modified iron oxide nanoparticles (IONP-PLL), which is formed by modifying poly-L-lysine to the surface of iron oxide nanoparticles. The ability of IONP-PLL to bind DNA was determined by ratio-dependent retardation of DNA in the agarose gel and co-sedimentation assay. In vitro cytotoxic effects were quantified by MTT assay. The transfection efficiency in vitro was evaluated by delivering exogenous DNA to different cell lines using IONP-PLL. Intravenous injection of IONP-PLL/DNA complexes into mice was evaluated as a gene delivery system for gene therapy. The PGL2-control gene encoding firefly luciferase and the EGFP-C2 gene encoding green fluorescent protein were used as marker genes. RESULTS IONP-PLL could bind and protect DNA. In contrast to PLL and cationic liposomes, IONP-PLL described here was less cytotoxic in a broad range of concentrations. In the current study, we have demonstrated that IONP-PLL can deliver exogenous gene to cells in vitro and in vivo. After intravenous injection, IONP-PLL transferred reporter gene EGFP-C2 to lung, brain, spleen and kidney. Furthermore, we have demonstrated that IONP-PLL transferred exogenous DNA across the blood-brain barrier to the glial cells and neuron of brain. CONCLUSIONS IONP-PLL, a low-toxicity vector, appears to have potential for fundamental research and genetic therapy in vitro and in vivo, especially for gene therapy of CNS disease.
Collapse
Affiliation(s)
- Juan-Juan Xiang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhao R, Gish K, Murphy M, Yin Y, Notterman D, Hoffman WH, Tom E, Mack DH, Levine AJ. The transcriptional program following p53 activation. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 65:475-82. [PMID: 12760064 DOI: 10.1101/sqb.2000.65.475] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- R Zhao
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Satoh T, Timme TL, Saika T, Ebara S, Yang G, Wang J, Ren C, Kusaka N, Mouraviev V, Thompson TC. Adenoviral vector-mediated mRTVP-1 gene therapy for prostate cancer. Hum Gene Ther 2003; 14:91-101. [PMID: 12614561 DOI: 10.1089/104303403321070793] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
We previously identified the mouse RTVP-1 (mRTVP-1; related to testes-specific, vespid, and pathogenesis proteins) gene as a direct target of p53 with proapoptotic activities in various cancer cell lines, including prostate cancer. To test the therapeutic potential of mRTVP-1 we constructed an adenoviral vector capable of efficient transduction and expression of mRTVP-1 (AdmRTVP-1) and used this vector in an orthotopic, metastatic mouse model of prostate cancer. A single intratumoral administration of AdmRTVP-1 gene therapy significantly reduced primary tumor wet weight compared with control Adbetagal-injected tumors at two time points after injection with two different vector doses (p < or = 0.01 at 7 and 14 days). Spontaneous metastasis to lung was also significantly reduced (p < or = 0.02). Evaluation of treated tumors revealed increased apoptosis and lower microvessel density counts. In a rat aortic ring sprouting assay, AdmRTVP-1 inhibited endothelial cell sprouting compared with Adbetagal, confirming its antiangiogenic activity. These therapeutic activities were associated with a significant increase in survival from 22.9 to 26.8 days (p = 0.003) in this aggressive model of prostate cancer. Interestingly, there were significant increases in the infiltration of tumor-associated macrophages, dendritic cells, and CD8+ T cells, which persisted at 14 days posttreatment in the AdmRTVP-1-treated tumors compared with Adbetagal control-treated tumors. In addition, significantly increased natural killer and cytotoxic T lymphocyte activities were demonstrated in the mice with AdmRTVP-1-treated tumors. The unique therapeutic properties of AdmRTVP-1 gene therapy demonstrated in this study provide new opportunities for gene and immunotherapy of prostate cancer and potentially other malignancies.
Collapse
Affiliation(s)
- Takefumi Satoh
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Nakashima Y, Yano M, Kobayashi Y, Moriyama S, Sasaki H, Toyama T, Yamashita H, Fukai I, Iwase H, Yamakawa Y, Fujii Y. Endostatin gene therapy on murine lung metastases model utilizing cationic vector-mediated intravenous gene delivery. Gene Ther 2003; 10:123-30. [PMID: 12571641 DOI: 10.1038/sj.gt.3301856] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumors require ongoing angiogenesis to support their growth. Inhibition of angiogenesis by production of antiangiogenic factors should be a viable approach for cancer gene therapy. In this study, we investigated whether intravenous administration of endostatin gene complexed with a cationic vector (GL67/DOPE or PEI22K) could inhibit the development of lung tumors in mice injected i.v. with NFSa Y83 fibrosarcoma cells (5 x 10(5)) which frequently form lung metastasis. mRNA and protein of the transfected gene were produced in the lung and other organs of the transfected mice as assessed by immunohistochemistry, Western blotting and reverse transcription-polymerase chain reaction. Single intravenous injection of the endostatin gene (60 microg) complexed with either GL67/DOPE or PEI22K on day 3 or day 7 after fibrosarcoma cell inoculation significantly inhibited tumor formation in the lung as evidenced by the reduced number of lung tumors and lung weight, and prolonged survival of the endostatin gene-transfected mice compared with control mice. These findings suggested that the endostatin gene therapy, using cationic vector-mediated intravenous gene transfer, might be a feasible strategy for organ-targeted prevention and regulation of possible disseminated cancers.
Collapse
Affiliation(s)
- Y Nakashima
- Department of Surgery II, Nagoya City University Medical School, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Nemunaitis J, Edelman J. Selectively replicating viral vectors. Cancer Gene Ther 2002; 9:987-1000. [PMID: 12522438 DOI: 10.1038/sj.cgt.7700547] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2002] [Indexed: 01/26/2023]
Affiliation(s)
- John Nemunaitis
- US Oncology, Inc., Collins Building, 5th Floor, Dallas, Texas 75246, USA.
| | | |
Collapse
|
31
|
Qiu ZH, Wu CT, Lao MF, Pan LZ, Li YM. Growth suppression and immunogenicity enhancement of Hep-2 or primary laryngeal cancer cells by adenovirus-mediated co-transfer of human wild-type p53, granulocyte-macrophage colony-stimulating factor and B7-1 genes. Cancer Lett 2002; 182:147-54. [PMID: 12048160 DOI: 10.1016/s0304-3835(02)00089-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Co-transfer of immunomodulatory and antiproliferative genes may be the basis for new strategies to potentiate tumor regression. In this study, we evaluated the in vitro effect of the introduction of human wild-type p53, granulocyte-macrophage colony-stimulating factor (GM-CSF), and B7-1 genes via recombinant adenovirus on the growth and immunogenicity of Hep-2 or primary laryngeal cancer cells. By the introduction of wild-type p53 gene, the growth of Hep-2 cells was inhibited via enhanced apoptosis. By the introduction of GM-CSF and B7-1 genes, the immunogenicity of cancer cells was enhanced. Significant proliferation of tumor infiltrating lymphocytes (TILs) and tumor-specific cytotoxicity of cytotoxic T lymphocytes (CTLs) were induced in vitro. Furthermore, the combinative effect of GM-CSF and B7-1 was even more evident than that of any one of them singly. These results suggest that the co-transfer of human wild-type p53, GM-CSF and B7-1 genes into tumor cells via recombinant adenovirus may be further developed into a potential combination gene therapy strategy for cancer.
Collapse
Affiliation(s)
- Zhao-hua Qiu
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, P.R. China
| | | | | | | | | |
Collapse
|
32
|
Dass CR. Biochemical and biophysical characteristics of lipoplexes pertinent to solid tumour gene therapy. Int J Pharm 2002; 241:1-25. [PMID: 12086717 DOI: 10.1016/s0378-5173(02)00194-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cationic liposomes have become the reagent of choice for transfer of nucleic acids such as plasmids and oligodeoxynucleotides to cells in culture and in vivo. Whilst these reagents have several advantages over other forms of nucleic acid transfer methods, toxicity remains a significant problem, especially in vivo. Recent studies have also highlighted the immunostimulatory nature of these cationic vesicles when complexed to plasmid DNA, a phenomenon that may be harnessed for efficacious usage against tumours. Current research in this dynamic technological field is aimed at the development of cationic lipids that have negligible toxic effects and enhanced transfection capabilities.
Collapse
Affiliation(s)
- Crispin R Dass
- Johnson & Johnson Research, 1 Central Avenue, Australian Technology Park, 1430, Eveleigh, Australia.
| |
Collapse
|
33
|
Kim SK, Wang KC, Cho BK, Chung HT, Kim YY, Lim SY, Lee CT, Kim HJ. Interaction between p53 and p16 expressed by adenoviral vectors in human malignant glioma cell lines. J Neurosurg 2002; 97:143-50. [PMID: 12134905 DOI: 10.3171/jns.2002.97.1.0143] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECT Multiple gene replacements have been examined as a potential treatment modality for malignant gliomas. Nevertheless, no reports are available that detail the synergy, additivity, or antagonism of multiple genes. The aim of this study was to assess the interaction between p53 and p16 genes in the growth of glioma cell lines. METHODS The human glioma cell lines U87MG and U373MG were transduced using an adenoviral vector with Ad-p53, Ad-p16, or both. Western blotting was performed to determine the expression of the protein products of the transduced p53 and p16 genes. To establish whether the combination of Ad-p53 and Ad-p16 would be beneficial, the effects of gene combinations at the median inhibitory concentration level were analyzed using the isobologram method. Annexin assays and cell cycle analyses were performed on the transduced cells. Western blotting demonstrated the expression of p53 and p16 in transduced cells. Simultaneous exposure to Ad-p53 and Ad-p16 produced additive effects in both glioma cell lines. Experimental data points in U373MG lay near the Mode I line, indicating that the vectors had a different mode of action. The restoration of normal p53-encoded protein in the mutant cell lines induced apoptosis, whereas in the wild-type p53 cell lines, the overexpression of wild-type p53 resulted in a moderate degree of apoptosis and G1 arrest. Furthermore, Ad-p16 induced more marked G1 arrest than Ad-p53 in cells with wild-type p53. CONCLUSIONS The results show that interaction between Ad-p53 and Ad-p16 is additive, regardless of p53 gene status.
Collapse
Affiliation(s)
- Seung-Ki Kim
- Department of Neurosurgery, Seoul National University, College of Medicine, Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Chen QR, Zhang L, Luther PW, Mixson AJ. Optimal transfection with the HK polymer depends on its degree of branching and the pH of endocytic vesicles. Nucleic Acids Res 2002; 30:1338-45. [PMID: 11884631 PMCID: PMC101355 DOI: 10.1093/nar/30.6.1338] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2001] [Revised: 01/22/2002] [Accepted: 01/22/2002] [Indexed: 11/14/2022] Open
Abstract
We have recently reported that liposomes in combination with histidine (HK)-containing polymers enhanced the expression of luciferase in transfected cells. In transformed or malignant cell lines, branched HK polymers (combined with liposome carriers) were significantly more effective than the linear HK polymer in stimulating gene expression. In the current study, we found that the linear HK polymer enhanced gene expression in primary cell lines more effectively than the branched polymers. The differences in the optimal carrier (linear versus branched) were not due to initial cellular uptake, size of the complexes or level of gene expression. There was, however, a strong association between the optimal type of HK polymer and the pH of endocytic vesicles (P = 0.0058). By altering the percentage of histidines carrying a positive charge, the endosomal pH of a cell may determine the amount of DNA released from the linear or branched HK polymer. In the two cell lines in which the linear HK was the optimal polymer, the endocytic vesicles were strongly acidic with a pH of <5.0. Conversely, in the four cell lines in which the branched polymers were optimal transfection agents, the pH of endocytic vesicles was >6.0. Furthermore, binding data support the relationship between DNA release from the optimal HK polymer and endosomal pH. The interplay between optimal HK polymers and the endosomal pH may lead to improved gene-delivery polymers tailored to a particular cell.
Collapse
Affiliation(s)
- Qing-Rong Chen
- Department of Pathology, University of Maryland Baltimore, MSTF Building, 10 South Pine Street, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
35
|
Nemunaitis J, Cunningham C. Emerging new therapies for chemotherapy-resistant cancer using adenoviral vectors. Drug Resist Updat 2002; 5:34-46. [PMID: 12127862 DOI: 10.1016/s1368-7646(02)00048-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The treatment of cancer by genetic manipulation of either the tumor itself or the patient as a whole offers new avenues for the treatment of otherwise refractory cancers. Gene therapy seeks to correct underlying genetic defects in malignant tissue or to augment the host defense response or to promote selectivity of other therapies. Many innovative and exciting genetic targets have been recently identified. However, the field as a whole is still constrained by limitations of gene delivery. The most common vector for gene delivery is modified adenovirus. In this review, we survey a sampling of current therapeutic approaches that depend upon adenoviral delivery vehicles and outline the advantages and disadvantages of this vector system.
Collapse
Affiliation(s)
- John Nemunaitis
- US Oncology, Mary Crowley Medical Research Center, Baylor University Medical Center, 3535 Worth Street, Collins Building, 5th Floor, Dallas, TX 75246, USA
| | | |
Collapse
|
36
|
Nemunaitis J, O'Brien J. Head and neck cancer: gene therapy approaches. Part 1: adenoviral vectors. Expert Opin Biol Ther 2002; 2:177-85. [PMID: 11849117 DOI: 10.1517/14712598.2.2.177] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Treatment options for recurrent or refractory head and neck cancer are limited. The goal of gene therapy is to introduce new genetic material into cancer cells without affecting toxicity to surrounding malignant cells. The most common vehicles for delivery of genes are adenoviruses. Adenoviruses gain access to malignant and normal cell cytoplasm via viral ligand binding to a unique cell surface receptor (the coxsackie adenovirus receptor [CAR]). However, this receptor is not cancer specific. Genetic modification of adenoviral DNA can create cancer specific targeting. Adenoviruses can be modified to express cancer specific ligands thereby focusing binding to malignant tissue. Furthermore, adenoviral delivered genes can be put under cancer specific promoter control to further limit gene expression in malignant tissue. Increased antitumour activity from such modifications has been demonstrated preclinically and several clinical trials have been completed demonstrating safety and clinical activity of non-replicating and conditional replicating adenoviral vector thereby opening the door for gene delivery and cancer specific targeting.
Collapse
Affiliation(s)
- John Nemunaitis
- 3535 Worth Street, Collins Building, 5th floor, Dallas, Texas 75246, USA.
| | | |
Collapse
|
37
|
Abstract
The vasculature of a tumour provides the most effective route by which neoplastic cells may be reached and eradicated by drugs. The fact that a tumour's vasculature is relatively more permeable than healthy host tissue should enable selective delivery of drugs to tumour tissue. Such delivery is relevant to carrier-mediated delivery of genetic medicine to tumours. This review discusses the potential of delivering therapeutic oligonucleotides (ONs) to tumours using cationic liposomes and cyclodextrins (CyDs), and the major hindrances posed by the tumour itself on such delivery. Cationic liposomes are generally 100-200 nm in diameter, whereas CyDs typically span 1.5 nm across. Cationic liposomes have been used for the introduction of nucleic acids into mammalian cells for more than a decade. CyD molecules are routinely used as agents that engender cholesterol efflux from lipid-laden cells, thus having an efficacious potential in the management of atherosclerosis. A recent trend is to employ these oligosaccharide molecules for delivering nucleic acids in cells both in-vitro and in-vivo. Comparisons are made with other ON delivery agents, such as porphyrin derivatives (< 1 nm), branched chain dendrimers (approximately 10 nm), polyethylenimine polymers (approximately 10 nm), nanoparticles (20-1,000 nm) and microspheres (> 1 microm), in the context of delivery to solid tumours. A discourse on how the chemical and physical properties of these carriers may affect the uptake of ONs into cells, particularly in-vivo, forms a major basis of this review.
Collapse
Affiliation(s)
- Crispin R Dass
- Johnson & Johnson Research, Strawberry Hills, Australia.
| |
Collapse
|
38
|
Sotomayor MG, Yu H, Antonia S, Sotomayor EM, Pardoll DM. Advances in gene therapy for malignant melanoma. Cancer Control 2002; 9:39-48. [PMID: 11907465 DOI: 10.1177/107327480200900106] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The recent developments in the field of gene transfer have advanced the use of gene therapy as a novel strategy against a variety of human malignancies. Due to its unique set of characteristics, melanoma represents a suitable target for the clinical translation of the different gene transfer approaches recently developed. The goal of gene therapy targeted to melanoma cells is to introduce "suicide" genes, to transfer tumor suppressor genes, to inactivate aberrant oncogene expression, or to introduce genes encoding immunologically relevant molecules. Gene therapy targeted to the host's immune cells has been developed as an additional strategy to redirect immune responses against melanoma. METHODS The authors reviewed the published gene transfer studies in experimental models, as well as the results of gene therapy clinical trials for patients with melanoma. RESULTS Clinical trials have shown the feasibility and safety of gene therapy against malignant melanoma. Although no major successes have been reported, the positive results observed in some patients support the potential for gene therapy in the management of this disease. CONCLUSIONS Gene therapy of melanoma using current gene transfer approaches is feasible and safe. Better vector technology as well as increased understanding of the "bystander effect" triggered by gene transfer approaches would provide the tools to validate gene therapy as an effective modality of treatment for malignant melanoma.
Collapse
Affiliation(s)
- Maria G Sotomayor
- Cutaneous Oncology Program, H. Lee Moffitt Cancer Center & Research Institute at the University of South Florida, Tampa, USA
| | | | | | | | | |
Collapse
|
39
|
Wysocki PJ, Mackiewicz-Wysocka M, Mackiewicz A. Cancer gene therapy – state-of-the-art. Rep Pract Oncol Radiother 2002. [DOI: 10.1016/s1507-1367(02)70989-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
40
|
Sherif ZA, Nakai S, Pirollo KF, Rait A, Chang EH. Downmodulation of bFGF-binding protein expression following restoration of p53 function. Cancer Gene Ther 2001; 8:771-782. [PMID: 11687900 DOI: 10.1038/sj.cgt.7700361] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2001] [Indexed: 12/23/2022]
Abstract
Angiogenesis is a requirement for solid tumor growth. Therefore, inhibition of this neovascularization is one mechanism by which restoration of wtp53 function may lead to tumor regression. Here we report that adenoviral vector-mediated wild-type p53 transduction results in growth inhibition of squamous cell carcinoma of the head and neck tumor cells both in vitro and in a xenograft mouse model. This growth inhibition is associated with the down-regulation of the expression of fibroblast growth factor binding protein, a secreted protein required for the activation of angiogenic factor basic FGF. These findings suggest that wtp53-induced tumor regression is due, at least in part, to antiangiogenesis mediated by the downmodulation of fibroblast growth factor binding protein.
Collapse
Affiliation(s)
- Z A Sherif
- Department of Oncology, Lombardi Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20007, USA
| | | | | | | | | |
Collapse
|
41
|
Densmore CL, Kleinerman ES, Gautam A, Jia SF, Xu B, Worth LL, Waldrep JC, Fung YK, T'Ang A, Knight V. Growth suppression of established human osteosarcoma lung metastases in mice by aerosol gene therapy with PEI-p53 complexes. Cancer Gene Ther 2001; 8:619-27. [PMID: 11593330 DOI: 10.1038/sj.cgt.7700343] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2001] [Indexed: 11/09/2022]
Abstract
Lung metastases are a frequent complication of osteosarcoma and a treatment that would reduce the severity of this complication would be of great benefit to patients. We have used a formulation consisting of polyethyleneimine (PEI) and a p53 gene administered in aerosol to treat established lung micrometastases as a model of human osteosarcoma in nude mice. The SAOS-LM6 cell line, a metastatic derivative of the p53 null SAOS-2 line, expresses high levels of p53 protein after in vitro transfection with PEI-p53 complexes as determined by ELISA, and transfection with both p53wt and the p53 variant, p53-CD(1-366) in vitro, results in a marked inhibition of SAOS-LM6 cell proliferation. Aerosol delivery of plasmid DNA containing either the p53 gene or a p53-CD(1-366) variant gene formulated with PEI to mice resulted in highly significant reductions in the numbers and size of tumors (P<.001), the total number of tumor foci in the lungs (P<.001) and the size of individual tumor nodules in treated animals compared to untreated, PEI only-treated and PEI-CAT-treated control animals. The different tissues examined did not reveal any signs of toxicity or inflammation after repeated exposure to PEI-DNA. The aerosol delivery of PEI-based formulations of p53 or synthetic p53 variant genes represents a promising new strategy for the treatment of established human osteosarcoma lung metastases. The noninvasive nature of aerosol delivery coupled with low toxicity also make this therapeutic approach potentially appropriate for combination therapy with either radio- or chemotherapy.
Collapse
Affiliation(s)
- C L Densmore
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The detection of free, circulating tumor DNA in the plasma of cancer patients opens up new possibilities for the diagnosis and prognostication of cancer. Whatever might be the mechanism for the presence of such DNA, it is now clear that oncogenes can circulate in the plasma fraction of blood and we can now ask whether this phenomenon has potentially important implications in cancer patients. The results of our experiments, together with previous observations of other authors, have led us to propose the "Hypothesis of Genometastasis", which suggests that metastases might develop as a result of transfection of susceptible cells in distant target organs with dominant oncogenes that circulate in the plasma and are derived from the primary tumor.
Collapse
|
43
|
Abstract
A recent target of cancer gene therapy is tumor angiogenesis. An appealing feature of gene therapy targeting the tumor vasculature is that it is readily accessible, particularly when the carrier and its gene are administered systemically. Several gene-based viral and nonviral therapies that target tumor angiogenesis have demonstrated the "proof of principle" of antiangiogenic therapy in preclinical models. The utility of antiangiogenic gene therapy in a clinical setting will depend in large part on developing vectors with minimal toxicity and with increased in vivo transfection efficiency. In this review, we discuss the current status and future directions of antiangiogenic gene therapy.
Collapse
Affiliation(s)
- Q R Chen
- Department of Pathology, University of Maryland at Baltimore, Baltimore, Maryland 21201, USA
| | | | | | | |
Collapse
|
44
|
Xu L, Pirollo KF, Chang EH. Tumor-targeted p53-gene therapy enhances the efficacy of conventional chemo/radiotherapy. J Control Release 2001; 74:115-28. [PMID: 11489488 DOI: 10.1016/s0168-3659(01)00324-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A long-standing goal in gene therapy for cancer is a stable, low toxic, systemic gene delivery system that selectively targets tumor cells, including metastatic disease. Progress has been made toward developing non-viral, pharmaceutical formulations of genes for in vivo human therapy, particularly cationic liposome-mediated gene transfer systems. Ligand-directed tumor targeting of cationic liposome-DNA complexes (lipoplexes) is showing promise for targeted gene delivery and systemic gene therapy. Lipoplexes directed by ligands such as folate, transferrin or anti-transferrin receptor scFv, showed tumor-targeted gene delivery and expression in human breast, prostate, head and neck cancers. The two elements, ligand/receptor and liposome composition, work together to realize the goal of functional tumor targeting of gene therapeutics. The tumor suppressor gene, p53, has been shown to be involved in the control of DNA damage-induced apoptosis. Loss or malfunction of this p53-mediated apoptotic pathway has been proposed as one mechanism by which tumors become resistant to chemotherapy or radiation. The systemically delivered ligand-liposome-p53 gene therapeutics resulted in efficient expression of functional wild-type p53, sensitizing the tumors to chemotherapy and radiotherapy. This is a novel strategy combining current molecular medicine with conventional chemotherapy and radiotherapy for the treatment of cancer. The systemic delivery of normal tumor suppressor gene p53 by a non-viral, tumor-targeted delivery system as a new therapeutic intervention has the potential to critically impact the clinical management of cancer.
Collapse
Affiliation(s)
- L Xu
- Department of Oncology, Lombardi Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | | | |
Collapse
|
45
|
Qiu Z, Lao M, Wu C. Co-transfer of human wild-type p53 and granulocyte-macrophage colony-stimulating factor genes via recombinant adenovirus induces apoptosis and enhances immunogenicity in laryngeal cancer cells. Cancer Lett 2001; 167:25-32. [PMID: 11323095 DOI: 10.1016/s0304-3835(01)00453-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Co-transfer of immunomodulatory and anti-proliferative genes may be the basis for new strategies to enhance tumor regression. The purpose of this study was to develop a combination gene therapy strategy for the treatment of laryngeal cancer. Human wild-type p53 and granulocyte-macrophage colony-stimulating factor (GM-CSF) genes were transferred into human laryngeal cancer cells mediated by adenovirus type 5 vector co-expressing human wild-type p53 and GM-CSF (Ad-p53/GM-CSF). By the introduction of the wild-type p53 gene, the growth of human laryngeal cancer Hep-2 cells was inhibited and their apoptosis was induced. By the introduction of the GM-CSF gene, the immunogenicity of cancer cells was enhanced. Significant proliferation of tumor infiltrating lymphocytes and tumor-specific cytotoxicity of cytotoxic T lymphocytes were induced by Ad-p53/GM-CSF-infected cancer cells in vitro. The results suggest that the co-transfer of human wild-type p53 and GM-CSF genes into tumor cells via recombinant adenovirus may be further developed into an effective and practical combination gene therapy strategy for laryngeal cancer.
Collapse
Affiliation(s)
- Z Qiu
- Beijing Institute of Radiation Medicine, 27 Taiping Road, 100850, Beijing, PR China
| | | | | |
Collapse
|
46
|
Slaton JW, Benedict WF, Dinney CP. P53 in bladder cancer: mechanism of action, prognostic value, and target for therapy. Urology 2001; 57:852-9. [PMID: 11337281 DOI: 10.1016/s0090-4295(01)00968-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- J W Slaton
- Departments of Urology and Genitourinary Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | | | | |
Collapse
|
47
|
Lebedeva S, Bagdasarova S, Tyler T, Mu X, Wilson DR, Gjerset RA. Tumor suppression and therapy sensitization of localized and metastatic breast cancer by adenovirus p53. Hum Gene Ther 2001; 12:763-72. [PMID: 11339893 DOI: 10.1089/104303401750148685] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have examined the effects of a replication-defective adenovirus encoding p53 (RPR/INGN 201 [Ad5CMV-p53]; Adp53), alone or in combination with the breast cancer therapeutic doxorubicin (Adriamycin), to suppress growth and induce apoptosis in breast cancer cells in vitro. We have also examined the in vivo effect of intratumoral administration of Adp53, alone or in combination with doxorubicin, to suppress the growth of established subcutaneous MDA-MB-435 breast cancer tumors. Finally, using the MDA-MB-435 orthotopic model of metastatic breast cancer, we have examined the effect of systemic administration of Adp53, alone or in combination with doxorubicin, to reduce the incidence of metastases. We find that whereas in vitro treatment of cells with Adp53 reduces [(3)H]thymidine incorporation by about 90% at 48 hr, cell viability at 6 days is reduced by only some 50% relative to controls. Although apoptosis is detectable in Adp53-treated cultures, these results suggest that a large fraction of Adp53-treated cells merely undergo reversible cell cycle arrest. Combined treatment with Adp53 and doxorubicin results in a greater than additive loss of viability in vitro and increased apoptosis. In vivo, locally administered Adp53 suppresses growth of established subcutaneous tumors in nude mice and suppression is enhanced by doxorubicin. In the metastatic breast cancer model, systemic administration of Adp53 plus doxorubicin leads to a significant reduction in the incidence of metastases relative to Adp53 or doxorubicin alone. Taken together, these data indicate an additive to synergistic effect of Adp53 and doxorubicin for the treatment of primary and metastatic breast cancer.
Collapse
Affiliation(s)
- S Lebedeva
- Sidney Kimmel Cancer Center, San Diego, CA 92121, USA
| | | | | | | | | | | |
Collapse
|
48
|
Chen QR, Zhang L, Stass SA, Mixson AJ. Branched co-polymers of histidine and lysine are efficient carriers of plasmids. Nucleic Acids Res 2001; 29:1334-40. [PMID: 11239000 PMCID: PMC29747 DOI: 10.1093/nar/29.6.1334] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We previously determined that a linear co-polymer of histidine and lysine (HK) in combination with liposomes enhanced the transfection efficiency of cationic liposomes. In the current study, we designed a series of HK polymers with increased branching and/or histidine/lysine ratio to determine if either variable affects transfection efficiency. In the presence of liposomes, the branched polymer with the highest number of histidines, HHK4b, was the most effective at enhancing gene expression. Furthermore, when serum was added to the medium during transfection, the combination of HHK4b and liposomes as a gene-delivery vehicle increased luciferase expression 400-fold compared to liposomes alone. In contrast to linear HK polymers, the higher branched HHK polymers were effective carriers of plasmids in the absence of liposomes. Without liposomes, the HHK4b carrier enhanced luciferase expression 15-fold in comparison with the lesser branched HHK2b carrier and increased expression by 5-logs in comparison with the HHK or HK carrier. The interplay of several parameters including increased condensation of DNA, buffering of acidic endosomes and differential binding affinities of polymer with DNA have a role in the enhancement of transfection by the HK polymers. In addition to suggesting that branched HK polymers are promising gene-delivery vehicles, this study provides a framework for the development of more efficient peptide-bond-based polymers of histidine and lysine.
Collapse
Affiliation(s)
- Q R Chen
- Department of Pathology and Greenebaum Cancer Center, University of Maryland Baltimore, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
49
|
Nemunaitis J, Khuri F, Ganly I, Arseneau J, Posner M, Vokes E, Kuhn J, McCarty T, Landers S, Blackburn A, Romel L, Randlev B, Kaye S, Kirn D. Phase II trial of intratumoral administration of ONYX-015, a replication-selective adenovirus, in patients with refractory head and neck cancer. J Clin Oncol 2001; 19:289-98. [PMID: 11208818 DOI: 10.1200/jco.2001.19.2.289] [Citation(s) in RCA: 308] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To determine the safety, humoral immune response replication, and activity of multiple intratumoral injections of ONYX-015 (replication selective adenovirus) in patients with recurrent squamous cell carcinoma of the head and neck (SCCHN). PATIENTS AND METHODS This phase II trial enrolled patients with SCCHN who had recurrence/relapse after prior conventional treatment. Patients received ONYX-015 at a dose of 2 x 10(11) particles via intratumoral injection for either 5 consecutive days (standard) or twice daily for 2 consecutive weeks (hyperfractionated) during a 21-day cycle. Patients were monitored for tumor response, toxicity, and antibody formation. RESULTS Forty patients (30 standard and 10 hyperfractionated) received 533 injections of ONYX-015. Standard treatment resulted in 14% partial to complete regression, 41% stable disease, and 45% progressive disease rates. Hyperfractionated treatment resulted in 10% complete response, 62% stable disease, and 29% progressive disease rates. Treatment-related toxicity included mild to moderate fever (67% overall) and injection site pain (47% on the standard regimen, 80% on the hyperfractionated regimen). Detectable circulating ONYX-015 genome suggestive of intratumoral replication was identified in 41% of tested patients on days 5 and 6 of cycle 1; 9% of patients had evidence of viral replication 10 days after injection during cycle 1, and no patients had evidence of replication > or = 22 days after injection. CONCLUSION ONYX-015 can be safely administered via intratumoral injection to patients with recurrent/refractory SCCHN. ONYX-015 viremia is transient. Evidence of modest antitumoral activity is suggested.
Collapse
|
50
|
Affiliation(s)
- M Shalev
- Matsunaga-Conte Prostate Cancer Research Center, Scott Department of Urology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|