Indian Journal of Human Genetics
Home Current Issue Archives Guidelines Subscriptions e-Alerts Login 
Users online: 18
Print this page  Email this page Small font sizeDefault font sizeIncrease font size


 
            Table of Contents  
REVIEW ARTICLE
Year : 2013  |  Volume : 19  |  Issue : 3  |  Page : 282-290
 

An overview of gene therapy in head and neck cancer


1 Department of Oral and Maxillofacial Surgery, M.M. College of Dental Sciences and Research, Mullana, Ambala, Haryana, India
2 Department of Periodontics, DAV (C) Dental College and Hospital, Yamuna Nagar, Haryana, India

Date of Web Publication30-Oct-2013

Correspondence Address:
Amit Bali
Department of Oral and Maxillofacial Surgery, M.M. College of Dental Sciences and Research, Mullana, Ambala, Haryana
India
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/0971-6866.120811

Get Permissions

 

   Abstract 

Gene therapy is a new treatment modality in which new gene is introduced or existing gene is manipulated to cause cancer cell death or slow the growth of the tumor. In this review, we have discussed the different treatment approaches for cancer gene therapy; gene addition therapy, immunotherapy, gene therapy using oncolytic viruses, antisense ribonucleic acid (RNA) and RNA interference-based gene therapy. Clinical trials to date in head and neck cancer have shown evidence of gene transduction and expression, mediation of apoptosis and clinical response including pathological complete responses. The objective of this article is to provide an overview of the current available gene therapies for head and neck cancer.


Keywords: Gene addition therapy, gene therapy, head and neck cancer, immunotherapy, suicide gene


How to cite this article:
Bali A, Bali D, Sharma A. An overview of gene therapy in head and neck cancer. Indian J Hum Genet 2013;19:282-90

How to cite this URL:
Bali A, Bali D, Sharma A. An overview of gene therapy in head and neck cancer. Indian J Hum Genet [serial online] 2013 [cited 2016 May 24];19:282-90. Available from: http://www.ijhg.com/text.asp?2013/19/3/282/120811



   Introduction Top


Conventional treatment modalities for head and neck cancer are surgery, radiotherapy and/or chemotherapy. These treatment modalities for head and neck cancer have not greatly improved the survival rate of patients. [1] Local and/or regional recurrence develops in approximately one-third of the patients despite definite treatment. [1] In patients with distant metastasis, recurrence is often considered as incurable. The failure to conventional therapy occurs because in case of extensively large tumors it may be difficult to clear the margins or some tumors are remarkably resistant to radiotherapy or chemotherapy. Unacceptable degree of toxicity and bystander damage to normal tissue occurs if we increase the dose of radiation or chemotherapeutic drug. [2] Combinations of current treatment modalities have been moderately successful, but often these combination therapies cause unacceptable toxicity without increasing the survival rate of patients. [2] The major drawback for this conventional treatment is a lack of specificity for the tumor cells and toxicity to the normal tissues.

Recent advances in molecular biology have documented the role of gene therapy in tumorogenesis. Gene therapy has potential to target cancer cells while sparing normal cell. The clinical application of the gene therapy for the treatment of head and neck cancer will require optimization of gene delivery in conjunction with determination of transfection efficiency. Gene therapy can be defined as "gene transfer for the purpose of treating human disease, this includes the transfer of new genetic material as well as manipulation of the existing genetic material." [1] In case of malignancy, with the help of gene therapy oncogenes can be targeted without damaging the normal cells. The aim of this article is to give you an overview of the current status of the gene therapy for head and neck cancer.


   Gene Therapy Approaches for Head and Neck Cancer Top


Targeting the specific genetic lesions responsible for carcinogenesis and cancer progression is an attractive strategy for developing more effective anticancer therapeutics and reducing treatment related toxicity. Any one of a battery of known genes expressed at the right site and at appropriate levels can be very effective for selectively killing cancer cells. Although systemic administration is theoretically desirable to address metastatic disease, gene therapy has not yet been shown to be suitable for systemic delivery in cancer patients. The head and neck cancer is an appropriate target for gene therapy because primary and recurrent lesions are readily accessible for injection or application of the agent. There are several general strategies utilized in a gene therapy approach to cancer, including.

Gene addition therapy

The regulation of tumor growth is achieved in this approach by introducing tumor suppressor genes that inactivate carcinogenic cells. [3] Cancer cells generally demonstrate impaired cell cycle progression, largely due to mutation and over expression of cell cycle regulators. [4] Several genetic alterations have been reported in the head and neck cancer including mutation of P 53 , the retinoblastoma gene (Rb1), P 16 and P 27 . Adult cells are normally maintained in either G 0 or G 1 stage of cell cycle by a signal from Rb pathway and P 53 pathway. [2] The tumor suppressor genes (P 53 , P 16 and P 27 ) and protein products of several proto-oncogenes plays a significant role in this pathway. Mutation in some of these genes may leads to cancer by eliminating tight control of cell proliferation. The Rb protein regulate the release from G 1 phase, whereas P 53 dictate whether cell cycle will be arrested in response to stress or deoxyribonucleic acid (DNA) damage or whether cell cycle will be directed to undergo apoptosis. [2] Since the protein P 53 plays an important role in cell cycle and in apoptosis, it was tested in patients with squamous cell carcinoma by injecting directly into the tumor with an adenoviral vector expressing wild type P 53 . Various randomized studies of adenoviral P 53 are presently underway to determine its role as a surgical adjuvant and in combination with DNA damaging agents. [5],[6],[7] P 27 gene was also found to inhibit the cell cycle of tumor cells. It acts by inducing apoptosis and triggering the suppression of tumor growth. Mutation of gene P 27 has been demonstrated to highly related with the appearance of tongue cancer. [8],[9],[10] Other tumor suppressor gene, which were used for gene therapy are Rb gene, melanoma differentiation associated gene-7. Their anti-tumor and pro-apoptotic response is similar to those of interleukin-24 (IL-24), a Th1 cytokines that triggers an anti-tumor and pro-apoptotic response in the immune system. [1],[11],[12],[13]

Cancer gene therapy using oncolytic viruses

One of the most promising gene therapy approaches is the use of viruses that replicate only in tumor cells, designated oncolytic viruses. [3] Viruses commonly used in gene therapy include, retroviruses, adenoviruses and herpes viruses. [14] The use of oncolytic viruses emerged from the discovery of adenoviruses lacking E1B, which do not grow in normal cells, but grew in cells without P 53 gene, one of the most common characteristics of the tumor cells. [15],[16]

Retroviruses are ribonucleic acid (RNA) viruses that undergo reverse transcription after infecting a cell, thereby producing double stranded DNA. DNA integrated into the host genome will pass copies of genes to all the subsequent generations. [17],[18] There are strategies such as ex vivo transduction of leukocytes with drug resistant genes, for which retrovirus could be well suited. Large multinational glioma trials has been performed with a murine retrovirus coding for herpes simplex virus type 1 thymidine kinase (HSV-1-TK) followed by intravenous ganciclovir treatment. This treatment modality has shown some evidence of efficacy. Herpes viruses were the among the first replication competent viruses utilized for cancer treatment. Most herpes virus vectors are developed from strains of HSV-1. [19] When HSV-1 infected a cell it replicate within the cell, causes cell lyses and infection of the surrounding cells. A replication - conditional mutant of HSV has been shown to elicit anti-tumor response in preclinical models of glioma and metastatic colon cancer. [19] Two vectors, G207 and NV1020 are currently in Phase 1 and Phase 2 trials for the treatment of oral cancer. G207 is mutated so that it has attenuated neurovirulence and cannot replicate in non-dividing cells. NV1020, a derivative originally used for vaccine studies, has multiple mutations including a deletion in TK region and a deletion across the long and short component of the genome and an insertion of the TK gene under the control of the α 4 promoters. [20],[21],[22],[23] Adenovirus is an oncolytic virus, which can be designed to replicate selectively in cancer cells and kill them by lysis. [1] The DNA of adenovirus does not integrate into the host genome and thus its effects are transient. Therefore, multiple administrations of the vectors are usually required, in addition infection of most dividing and quiescent cell types occurs with unparalleled efficiency and the genome can accommodate relatively larger payloads. [1] The most notable adenoviral therapy is the ONYX-015 viral therapy. [24] Adenovirus ONYX-015 has been engineered to lack the viral E1B protein. In the absence of E1B protein, the virus is unable to replicate in healthy cell with a normal P 53 pathways. Due to mutations, cancer cells often have a deficiency in P 53 pathway and thus allow ONYX-015 to replicate and lyses the cell. ONYX-015 has been tested in Phase 1 and 2 trials on squamous cell carcinoma of head and neck that resulted in tumor regression, which correlate to the P 53 status of the tumor. Phase 2 trials of ONYX-015 in combination with chemotherapy has demonstrated even better response and have led to a Phase 3 study. [25],[26],[27],[28],[29] The presence of cytokines, IL-6, tumor necrosis factor-α (TNF-α), IL-10 and interferon-gamma (IFN-ϒ) is observed at 24 h after ONYX-015 administration. IFN-ϒ level increased after 4 days while TNF-α level increased after 6 h, thereby contributing toward the appearance of immune response to the tumor. [30],[31] Researchers have reported on the anti-tumor efficiency of the intravenous administration of oncolytic adenovirus OAS403. In 7-10 days, cytotoxicity appeared in tumor cells. This adenovirus is active in tumor with abnormal Rb protein and abnormality in the regulation of telomerase expression. The combination of OAS403 with conventional chemotherapeutic agent increased the anti-tumor efficacy in pre-clinical studies and Phase 3 trials are now underway. [32],[33],[34],[35] Ongoing clinical trials on oncolytic viral therapy are given in [Table 1].
Table 1: Ongoing trials of oncolytic virus therapy for head and neck cancer

Click here to view


Immunotherapy

In the immunotherapy either the immunogenic potential of tumor cells is increased and/or the patient's immune response to a tumor is augmented. Immunotherapy has been tried for cancer treatment for over 100 years. [1] However limited success has been achieved as cancer cells tend to evolve mechanism that evades immune detection. [36],[37],[38] Patients with head and neck squamous cell carcinoma have demonstrated functional deficiencies in several categories of immune cells including natural killer (NK) cells, T-lymphocytes and several cytokines. Animal studies have shown that IL-2 administration activate T-lymphocytes and NK cells that in turn activate TNF-α, triggered by strong tumor inhibition effect. [39] Mechanisms to increase the sensitivity of the tumor to normal therapeutic processes are under investigation. Suppression of nuclear factor kappa beta activates the anti-apoptotic proteins, TNF, TNF receptor-associated factor-1 (TRAF-1), TRAF-2, cellular inhibitor of apoptosis protein-1. [40],[41] Currently, gene therapy is being used to create recombinant cancer vaccine. These vaccines are not meant to prevent disease, but cure or curtain it by training the patient's immune system to recognize the cancer cells. There are different types of cancer vaccines at the different phase of clinical trials [42] [Table 2] and [Table 3]. Advances in the understanding of the mechanism of action of cellular anti-tumor immune response have allowed the development of a new generation of cancer vaccines. The most immunologically active vaccine usually required costly and laborious ex vivo cellular cultures, whereas the cell free vaccine that can be directly administered from an easily stored and transported vials are usually less immunologically active. New advances and precise knowledge of the requirement for the generation of cellular immune response to tumor antigen will likely provide powerful, non-individualized cell free vaccine in the near future. [42],[43],[44]
Table 2: Ongoing trials of gene immunotherapy for head and neck cancer

Click here to view
Table 3: Cancer vaccines

Click here to view


Suicide gene therapy

Suicide gene therapy is also termed as gene directed enzyme pro-drug therapy and this approach has attracted increasing attention. The basic principle behind the suicide gene therapy is the selective intratumoral activation of a non-toxic drug by specific transfer of the activating transgene into tumor cells. [45] This approach encompasses several therapeutic systems, but HSV-TK is most extensively utilized. This gene encodes a viral enzyme that phosphorylates ganciclovir into a monophosphate form, which is then phosphorylated by intracellular enzyme into an active triphosphate compound that terminate DNA synthesis. [46],[47] Suicide gene therapy can also use adenovirus. Adeno-associated virus (AAV) mediated delivery of the HSV-TK gene can selectively kill α-fetoprotein positive hepatocellular carcinoma cells in a mouse model and a bystander effect was demonstrated followed by the administration of the ganciclovir. [34],[48] Similar in vivo therapeutic effect of AAV-mediated delivery of HSV-TK gene has also been reported in an experimental glioma model and a human squamous cell carcinoma. [49],[50] Considerable knowledge is now available on genes that contribute toward resistance against chemotherapy, including multidrug resistance protein 1 (MDR1), multidrug related protein, dihydrofolate reductase. [35] MDR is associated with over expression of the P-glycoprotein. It is a 170-kDa transmembrane ATPase that export chemotherapeutic drug from cells. [51],[52] Li et al. has used self-complementary vectors scAAV to successfully deliver hairpin small interfering RNA (siRNA) into MDR human breast cancer and oral cancer cells. [52] It dramatically reduces P-glycoprotein expression levels resulting in substantial reversal of the MDR phenotype in the cells. One of the major drawbacks of the suicide gene therapy is transfection efficiency. [1] However, a high percentage of transfected cells do not appear to be required in vivo due to the ability of transfected tumor cells to induce cell death in neighboring transfected cells (bystander effect). [1] Ongoing clinical trials on suicide gene therapy are given in [Table 4].
Table 4: Ongoing trials of suicide gene therapy for head and neck cancer

Click here to view



   Antisense RNA Top


The activity of several known oncogenes including myc, fos and ras and certain viruses such as HSV-1, human papillomavirus and human T-lymphotropic virus type I can be inhibited by antisense RNA. [3] Antisense RNA is a complimentary strand of the DNA. Gene expression can usually be inhibited by the RNA. [1] Inhibition of expression of theses oncogenes may alter the phenotype, thus abrogating tumor growth. [1] Conventional use of this technique is limited by the difficulty of introducing a sufficient quantity of antisense molecules to inhibit tumor growth. To overcome this drawback, powerful promoters are being used. [53] Preclinical studies using antisense RNA promoters demonstrated a powerful anti-tumor effect with minimal toxicity. [54] A phase-1 study in patients with advanced oral cancer is underway to determine the safety and biological effect of liposome mediated intratumoral epidermal growth factor receptor antisense gene therapy. Results have been positive, showing a low toxicity and high efficacy. [54]


   RNA Interference (RNAi)-Based Gene Therapy Top


siRNA are mainly involved in guiding messenger RNA degradation. RNAi-based gene therapy consist of two approaches: Plasmid or viral vector mediated delivery of short hairpin RNA precursors and direct delivery of siRNAs or siRNA precursors to target cells. [55] RNAi-based gene therapy has been used to treat age related molecular degeneration and respiratory syncytial viral infection. [55] The application of RNAi-based gene therapy for cancer is in the preclinical stage. Although RNAi-based gene therapy has been confirmed efficacious, improvements are required and a number of challenges must be addressed to realize its full potential.


   Conclusion Top


As shown in this review, cancer gene therapy has been shown to be effective in initial clinical trials. As our understanding of the molecular mechanism of cancer increases, it is possible to exploit these principles to target tumor cells selectively. Very soon it may contribute a definitive treatment for head and neck cancers that will offer great effectiveness compared with current therapies and will markedly reduce the high mortality associated with these lesions. Gene therapy is now moving from Phase 1 and 2 trials to the next level of Phase 3 and 4 trials, but it will take considerable time and require a large number of patients to demonstrate the true efficacy of the therapies. Moreover, gene therapy is likely to be very effective in combination with pre-existing clinical regimen such as radiotherapy, chemotherapy and surgery. A large number of studies are now showing great potential for combining gene therapy and chemotherapeutic, immunological and radiotherapeutic approaches to kill cells more effectively and in greater numbers. The field of cancer gene therapy is rapidly maturing and will definitely be part of the future cancer therapeutics. While not all the current trials will lead to a variable therapeutic agent, there is greater hope that these advances will help to treat cancer patients without offering suffering and death.

 
   References Top

1.Xi S, Grandis JR. Gene therapy for the treatment of oral squamous cell carcinoma. J Dent Res 2003;82:11-6.  Back to cited text no. 1
    
2.Moon C, Oh Y, Roth JA. Current status of gene therapy for lung cancer and head and neck cancer. Clin Cancer Res 2003;9:5055-67.  Back to cited text no. 2
    
3.Ayllón Barbellido S, Campo Trapero J, Cano Sánchez J, Perea García MA, Escudero Castaño N, Bascones Martínez A. Gene therapy in the management of oral cancer: Review of the literature. Med Oral Patol Oral Cir Bucal 2008;13:E15-21.  Back to cited text no. 3
    
4.Cross D, Burmester JK. Gene therapy for cancer treatment: Past, present and future. Clin Med Res 2006;4:218-27.  Back to cited text no. 4
    
5.Cardinali M, Jakus J, Shah S, Ensley JF, Robbins KC, Yeudall WA. p21(WAF1/Cip1) retards the growth of human squamous cell carcinomas in vivo. Oral Oncol 1998;34:211-8.  Back to cited text no. 5
    
6.Clayman GL. The current status of gene therapy. Semin Oncol 2000;27 Suppl 8:39-43.  Back to cited text no. 6
    
7.Clayman GL, Trapnell BC, Mittereder N, Liu TJ, Eicher S, Zhang S, et al. Transduction of normal and malignant oral epithelium by an adenovirus vector: The effect of dose and treatment time on transduction efficiency and tissue penetration. Cancer Gene Ther 1995;2:105-11.  Back to cited text no. 7
    
8.Supriatno, Harada K, Kawaguchi S, Onoue T, Yoshida H, Sato M. Characteristics of antitumor activity of mutant type p27Kip1 gene in an oral cancer cell line. Oral Oncol 2004;40:679-87.  Back to cited text no. 8
    
9.Galanis E, Okuno SH, Nascimento AG, Lewis BD, Lee RA, Oliveira AM, et al. Phase I-II trial of ONYX-015 in combination with MAP chemotherapy in patients with advanced sarcomas. Gene Ther 2005;12:437-45.  Back to cited text no. 9
    
10.Liu X, Huang H, Wang J, Wang C, Wang M, Zhang B, et al. Dendrimers-delivered short hairpin RNA targeting hTERT inhibits oral cancer cell growth in vitro and in vivo. Biochem Pharmacol 2011;82:17-23.  Back to cited text no. 10
    
11.Shi H, Wang J, Dong F, Wang X, Li H, Hou Y. The effect of proteoglycans inhibited by RNA interference on metastatic characters of human salivary adenoid cystic carcinoma. BMC Cancer 2009;9:456.  Back to cited text no. 11
    
12.Yang Y, Ge PJ, Jiang L, Li FL, Zhu QY. Modulation of growth and angiogenic potential of oral squamous carcinoma cells in vitro using salvianolic acid B. BMC Complement Altern Med 2011;11:54.  Back to cited text no. 12
    
13.Uchida D, Onoue T, Begum NM, Kuribayashi N, Tomizuka Y, Tamatani T, et al. Vesnarinone downregulates CXCR4 expression via upregulation of Krüppel-like factor 2 in oral cancer cells. Mol Cancer 2009;8:62.  Back to cited text no. 13
    
14.Chahlavi A, Rabkin S, Todo T, Sundaresan P, Martuza R. Effect of prior exposure to herpes simplex virus 1 on viral vector-mediated tumor therapy in immunocompetent mice. Gene Ther 1999;6:1751-8.  Back to cited text no. 14
    
15.Heise C, Kirn DH. Replication-selective adenoviruses as oncolytic agents. J Clin Invest 2000;105:847-51.  Back to cited text no. 15
    
16.Wodarz D. Gene therapy for killing p53-negative cancer cells: Use of replicating versus nonreplicating agents. Hum Gene Ther 2003;14:153-9.  Back to cited text no. 16
    
17.Høgset A, Engesaeter BO, Prasmickaite L, Berg K, Fodstad O, Maelandsmo GM. Light-induced adenovirus gene transfer, an efficient and specific gene delivery technology for cancer gene therapy. Cancer Gene Ther 2002;9:365-71.  Back to cited text no. 17
    
18.Williams MD. Integration of biomarkers including molecular targeted therapies in head and neck cancer. Head Neck Pathol 2010;4:62-9.  Back to cited text no. 18
    
19.Maeda N, Kawamura T, Hoshino H, Yamada N, Blackard J, Kushida S, et al. Inhibition of human T-cell leukemia virus type 1 replication by antisense env oligodeoxynucleotide. Biochem Biophys Res Commun 1998;243:109-12.  Back to cited text no. 19
    
20.Cozzi PJ, Malhotra S, McAuliffe P, Kooby DA, Federoff HJ, Huryk B, et al. Intravesical oncolytic viral therapy using attenuated, replication-competent herpes simplex viruses G207 and Nv1020 is effective in the treatment of bladder cancer in an orthotopic syngeneic model. FASEB J 2001;15:1306-8.  Back to cited text no. 20
    
21.Yoon SS, Nakamura H, Carroll NM, Bode BP, Chiocca EA, Tanabe KK. An oncolytic herpes simplex virus type 1 selectively destroys diffuse liver metastases from colon carcinoma. FASEB J 2000;14:301-11.  Back to cited text no. 21
    
22.Hemminki A, Kanerva A, Kremer EJ, Bauerschmitz GJ, Smith BF, Liu B, et al. A canine conditionally replicating adenovirus for evaluating oncolytic virotherapy in a syngeneic animal model. Mol Ther 2003;7:163-73.  Back to cited text no. 22
    
23.Lujan B, Hakim S, Moyano S, Nadal A, Caballero M, Diaz A, et al. Activation of the EGFR/ERK pathway in high-grade mucoepidermoid carcinomas of the salivary glands. Br J Cancer 2010;103:510-6.  Back to cited text no. 23
    
24.Bischoff JR, Kirn DH, Williams A, Heise C, Horn S, Muna M, et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science 1996;274:373-6.  Back to cited text no. 24
    
25.O'Shea CC, Soria C, Bagus B, McCormick F. Heat shock phenocopies E1B-55K late functions and selectively sensitizes refractory tumor cells to ONYX-015 oncolytic viral therapy. Cancer Cell 2005;8:61-74.  Back to cited text no. 25
    
26.Nemunaitis J, Ganly I, Khuri F, Arseneau J, Kuhn J, McCarty T, et al. Selective replication and oncolysis in p53 mutant tumors with ONYX-015, an E1B-55kD gene-deleted adenovirus, in patients with advanced head and neck cancer: A phase II trial. Cancer Res 2000;60:6359-66.  Back to cited text no. 26
    
27.Lamont JP, Nemunaitis J, Kuhn JA, Landers SA, McCarty TM. A prospective phase II trial of ONYX-015 adenovirus and chemotherapy in recurrent squamous cell carcinoma of the head and neck (the Baylor experience). Ann Surg Oncol 2000;7:588-92.  Back to cited text no. 27
    
28.Rudin CM, Cohen EE, Papadimitrakopoulou VA, Silverman S Jr, Recant W, El-Naggar AK, et al. An attenuated adenovirus, ONYX-015, as mouthwash therapy for premalignant oral dysplasia. J Clin Oncol 2003;21:4546-52.  Back to cited text no. 28
    
29.Liu R, Varghese S, Rabkin SD. Oncolytic herpes simplex virus vector therapy of breast cancer in C3(1)/SV40 T-antigen transgenic mice. Cancer Res 2005;65:1532-40.  Back to cited text no. 29
    
30.Nemunaitis J, Cunningham C, Tong A, Post L, Netto G, Paulson AS, et al. Pilot trial of intravenous infusion of a replication-selective adenovirus (ONYX-015) in combination with chemotherapy or IL-2 treatment in refractory cancer patients. Cancer Gene Ther 2003;10:341-52.  Back to cited text no. 30
    
31.Imboden M, Shi F, Pugh TD, Freud AG, Thom NJ, Hank JA, et al. Safety of interleukin-12 gene therapy against cancer: A murine biodistribution and toxicity study. Hum Gene Ther 2003;14:1037-48.  Back to cited text no. 31
    
32.Manchanda P, Sharma SC, Das SN. Differential regulation of IL-2 and IL-4 in patients with tobacco-related oral squamous cell carcinoma. Oral Dis 2006;12:455-62.  Back to cited text no. 32
    
33.Fukui T, Hayashi Y, Kagami H, Yamamoto N, Fukuhara H, Tohnai I, et al. Suicide gene therapy for human oral squamous cell carcinoma cell lines with adeno-associated virus vector. Oral Oncol 2001;37:211-5.  Back to cited text no. 33
    
34.Neves SS, Sarmento-Ribeiro AB, Simões SP, Pedroso de Lima MC. Transfection of oral cancer cells mediated by transferrin-associated lipoplexes: Mechanisms of cell death induced by herpes simplex virus thymidine kinase/ganciclovir therapy. Biochim Biophys Acta 2006;1758:1703-12.  Back to cited text no. 34
    
35.Young M, Overlid N, Konopka K, Düzgünes N. Gene therapy for oral cancer: Efficient delivery of a 'suicide gene' to murine oral cancer cells in physiological milieu. J Calif Dent Assoc 2005;33:967-71.  Back to cited text no. 35
    
36.Rao SK, Pavicevic Z, Du Z, Kim JG, Fan M, Jiao Y, et al. Pro-inflammatory genes as biomarkers and therapeutic targets in oral squamous cell carcinoma. J Biol Chem 2010;285:32512-21.  Back to cited text no. 36
    
37.Kowalczyk DW, Wysocki PJ, Mackiewicz A. Cancer immunotherapy using cells modified with cytokine genes. Acta Biochim Pol 2003;50:613-24.  Back to cited text no. 37
    
38.Liu M, Acres B, Balloul JM, Bizouarne N, Paul S, Slos P, et al. Gene-based vaccines and immunotherapeutics. Proc Natl Acad Sci U S A 2004;101 Suppl 2:14567-71.  Back to cited text no. 38
    
39.Li D, Shugert E, Guo M, Bishop JS, O'Malley BW Jr. Combination nonviral interleukin 2 and interleukin 12 gene therapy for head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 2001;127:1319-24.  Back to cited text no. 39
    
40.Ramadoss S, Li J, Ding X, Al Hezaimi K, Wang CY. Transducin β-like protein 1 recruits nuclear factor κB to the target gene promoter for transcriptional activation. Mol Cell Biol 2011;31:924-34.  Back to cited text no. 40
    
41.Fang L, Hu Q, Hua Z, Li S, Dong W. Growth inhibition of a tongue squamous cell carcinoma cell line (Tca8113) in vitro and in vivo via siRNA-mediated down-regulation of skp2. Int J Oral Maxillofac Surg 2008;37:847-52.  Back to cited text no. 41
    
42.Clay TM, Hobeika AC, Mosca PJ, Lyerly HK, Morse MA. Assays for monitoring cellular immune responses to active immunotherapy of cancer. Clin Cancer Res 2001;7:1127-35.  Back to cited text no. 42
    
43.Ziai J, French CA, Zambrano E. NUT gene rearrangement in a poorly-differentiated carcinoma of the submandibular gland. Head Neck Pathol 2010;4:163-8.  Back to cited text no. 43
    
44.Schuurhuis DH, Ioan-Facsinay A, Nagelkerken B, van Schip JJ, Sedlik C, Melief CJ, et al. Antigen-antibody immune complexes empower dendritic cells to efficiently prime specific CD8+CTL responses in vivo. J Immunol 2002;168:2240-6.  Back to cited text no. 44
    
45.Kalergis AM, Ravetch JV. Inducing tumor immunity through the selective engagement of activating Fcgamma receptors on dendritic cells. J Exp Med 2002;195:1653-9.  Back to cited text no. 45
    
46.Lo WL, Yu CC, Chiou GY, Chen YW, Huang PI, Chien CS, et al. MicroRNA-200c attenuates tumour growth and metastasis of presumptive head and neck squamous cell carcinoma stem cells. J Pathol 2011;223:482-95.  Back to cited text no. 46
    
47.Yoo GH, Moon J, Leblanc M, Lonardo F, Urba S, Kim H, et al. A phase 2 trial of surgery with perioperative INGN 201 (Ad5CMV-p53) gene therapy followed by chemoradiotherapy for advanced, resectable squamous cell carcinoma of the oral cavity, oropharynx, hypopharynx, and larynx: Report of the Southwest Oncology Group. Arch Otolaryngol Head Neck Surg 2009;135:869-74.  Back to cited text no. 47
    
48.Mizuno M, Yoshida J, Colosi P, Kurtzman G. Adeno-associated virus vector containing the herpes simplex virus thymidine kinase gene causes complete regression of intracerebrally implanted human gliomas in mice, in conjunction with ganciclovir administration. Jpn J Cancer Res 1998;89:76-80.  Back to cited text no. 48
    
49.Kanazawa T, Urabe M, Mizukami H, Okada T, Kume A, Nishino H, et al. Gamma-rays enhance rAAV-mediated transgene expression and cytocidal effect of AAV-HSVtk/ganciclovir on cancer cells. Cancer Gene Ther 2001;8:99-106.  Back to cited text no. 49
    
50.Kanazawa T, Mizukami H, Nishino H, Okada T, Hanazono Y, Kume A, et al. Topoisomerase inhibitors enhance the cytocidal effect of AAV-HSVtk/ganciclovir on head and neck cancer cells. Int J Oncol 2004;25:729-35.  Back to cited text no. 50
    
51.Yuan Y, Liao YM, Hsueh CT, Mirshahidi HR. Novel targeted therapeutics: Inhibitors of MDM2, ALK and PARP. J Hematol Oncol 2011;4:16.  Back to cited text no. 51
    
52.Xu D, McCarty D, Fernandes A, Fisher M, Samulski RJ, Juliano RL. Delivery of MDR1 small interfering RNA by self-complementary recombinant adeno-associated virus vector. Mol Ther 2005;11:523-30.  Back to cited text no. 52
    
53.Bertrand JR, Pottier M, Vekris A, Opolon P, Maksimenko A, Malvy C. Comparison of antisense oligonucleotides and siRNAs in cell culture and in vivo. Biochem Biophys Res Commun 2002;296:1000-4.  Back to cited text no. 53
    
54.Yang L, Ma X, Xiao L, Tang M, Weng X, Sun L, et al. Uniquely modified RNA oligonucleotides targeting STAT3 suppress melanoma growth both in vitro and in vivo. Cancer Biol Ther 2009;8:2065-72.  Back to cited text no. 54
    
55.Zhang C, Wang QT, Liu H, Zhang ZZ, Huang WL. Advancement and prospects of tumor gene therapy. Chin J Cancer 2011;30:182-8.  Back to cited text no. 55
    



 
 
    Tables

  [Table 1], [Table 2], [Table 3], [Table 4]


This article has been cited by
1 Emerging drugs for head and neck cancer
Yihui Wen,Jennifer R Grandis
Expert Opinion on Emerging Drugs. 2015; : 1
[Pubmed] | [DOI]



 

Top
Print this article  Email this article
           

    

 
   Search
 
  
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Article in PDF (267 KB)
    Citation Manager
    Access Statistics
    Reader Comments
    Email Alert *
    Add to My List *
* Registration required (free)  


    Abstract
   Introduction
    Gene Therapy App...
   Antisense RNA
    RNA Interference...
   Conclusion
    References
    Article Tables

 Article Access Statistics
    Viewed1687    
    Printed35    
    Emailed1    
    PDF Downloaded75    
    Comments [Add]    
    Cited by others 1    

Recommend this journal