Open access

IGF-I Antisense and Triple-Helix Gene Therapy of Glioblastoma

Written By

Jerzy Trojan and Ignacio Briceno

Submitted: 08 August 2012 Published: 27 February 2013

DOI: 10.5772/52366

From the Edited Volume

Evolution of the Molecular Biology of Brain Tumors and the Therapeutic Implications

Edited by Terry Lichtor

Chapter metrics overview

1,642 Chapter Downloads

View Full Metrics

1. Introduction

1.1. General concept

There is a convergence between ontogenesis and carcinogenesis. In theory all antigens correspond to a specific stage of embryo/fetal development. These antigens – oncodevelopmental proteins - are also present in homologous neoplatic tissues during cancer growth. The first illustration of this theory is alpha-fetoprotein (AFP), a common tumor marker, which is present during the normal development of central nervous system of the rat. Immunohistochemical analysis of the localization of AFP during normal development of the central nervous system and in pathological homologous nervous tissues of teratocarcinoma, shows a remarkable parallelis [1]. A similar demonstration was demonstrated for serum albumin antigen and blood group H antigen [2]. As AFP was a specific marker for neuronal cells, a search for a specific marker of glial cells has constituted the next step in the nervous system research to distinguish glial and neuronal cells, because the known markers were not totally specific. It was demonstrated that the growth factor IGF-I (Insulin like Grow Factor I) [3-6] is present only in glial cells, and absent in neuronal cells in normal and neoplastic developmen [7]. IGF-I is, therefore, a specific marker of glioblastoma. The use of IGF-I for the study of malignant tumors, including the gliomas, became the best choice as a marker [8-10].

1.2. Anti — Gene strategies

1.2.1. Antisense

For nearly twenty years various treatments of tumors, based on the injection of antibodies directed against specific antigens and stem cells have been tested. The results were not successful because of non-selective affinity for the tumor. Moreover, direct injection of protein coupled to an isotope such as iodine-125, has also posed the problem of the limited specificity of the distribution of the protein [11]. After that, the researchers have approached the study in a radically different way, trying to stop the production of IGF-I, acting directly on the messenger RNA [7,12].

Since 1978 it is known that antisense messengers are naturally produced and destroyed in the process of DNA replicatio [13-17]. The authors successfully demonstrated that this phenomenon is possible in twenty different species. Fortunately, the researchers could produce artificial antisense messengers in a significant number [12,17-19]. Moreover, using antisense oligonucleotides associated with the photoluminescent amino-terminated poly amido amine dendrimer, they can be directly analyzed by fluorescence microscopy and flow cytometry [20]. The antisense sequences are capable of blocking the messenger RNA translation conducting to block of specific protein synthesis.

1.2.2. Triple helix

Since the 90s, another approach in parallel with the strategy of antisense RNA has become successful in gene therapy and clinical trials: the triple helix strategy [21-23]. The triple helix technology (HT) is the newest approach, which belongs to the antisense approach and sensu lato anti gene strategies. TH technology was discovered by PB Derwan and C. Helene [24,25] and its action was defined as inhibition of gene expression at the level of transcription. In short, specific oligonucleotide sequences (also called triple helix-forming oligonucleotides, TFOs) are introduced into cells by transfection using chemical carriers, such as plasmid vectors which can direct synthesis TFOs. The TFOs are linked to the genomic DNA forming the triple helix structure with the target gene and inhibiting its transcription. The TFOs usually are directed against sequences located in the promoter region of genes of interest [24]. Examples of the inhibitory activity of TFOs on target genes involved in tumorigenesis are currently available. This approach, based on the triple helix of DNA has been used for the inhibition of IGF-I, which plays a major role in tumorigenesis [26]. Triplex strategy has also been applied to the inhibition of Ras oncogenes which are the most frequently activated in human cancer. Transcription was inhibited by human Hras TFOs targeting sequences recognized by the transcription factor Sp I [27]. Moreover, synthesis of Human Tumor Necrosis Factor (TNF), which acts as an autocrine growth factor in various tumor cell lines including neuroblastoma and glioblastoma has been blocked by treatment with TFOs [28,29].

Advertisement

2. Experimental results

2.1. In vitro and in vivo experiments on glioma tumors

The vector of antisense type was transfected into established glioma C6 cell line. All transfected cells have changed phenotype. Morphologically these cells had an elongated appearance. Transfected glial cells have produced a large amount of antisense RNA [7,30].

Regarding in vivo experiment, subcutaneous injection of C6 glioma cells into DBX rats have produced a glioma tumor after ten days. When transfected glioma cells were injected (with vector carrying the cDNA antisense against IGF-I) the tumors did not develop.

The experience was reproducible in 200 rats and the results were confirmatory. It was demonstrated that if injected initially native C6 glioma cells were followed a week later by injection into another point of subcutaneously transfected glial cells, the tumor developed, but disappeared completely after 2 to 3 weeks [12].

The tumor examined histologically shortly before the death of animals, have shown an important lymphocytes / plasmocytes infiltration. Histological analysis of the tumor during its disappearance suggests that the essential mechanism is related to the presence of specifically CD8 + cytotoxic T lymphocytes. Systematic analysis of all tissues showed that these lymphocytes were of splenic origin, and were observed in very large quantities in the spleen of experimental animals (10 to15%) [12,31,32].

2.2. Mechanism of glioma tumor destruction

The cytotoxic T CD8+ cell can exert its effect, if a bridge between CD8 and the antigen of class I major histocompatibility complex (MHC) occurs [33-36]. Following transfection of glial cells with the antisense cDNA of IGF-I, the expression of MHC-I in transfected glioma cells and in vivo, is greatly enhanced (5 times). This mechanism may play a role in the cytotoxic response, although not the only one involve [37]. The bearing tumor rats receiving injections of transfected cells, have revealed a very high rate of CD8 + cells. This anti-tumor immune response has stopped the tumor development [12,31,38-40].

Advertisement

3. Application in clinical trial

3.1. Ethical committees

Approval for the clinical trial of gene therapy (based on the NIH clinical study No 1602, Bethesda, Maryland, 24. 11. 1993) [41] was administered by the Commission of Bioethics at the University of Medicine, Bromberg (No KB / 176/2001, 28. 06. 2002) and registered by Wiley International Gene Therapy Clinical Trial database No. 635 and 636 (J Gene Med, updated 2002), and by NATO Science program 2003/2007 - USA, France, Poland, Germany (LST 980 517).

3.2. Conditions

The NIH Committee raised a number of conditions. Initially, for all patients, tumor cells must be isolated from biopsies and cells clones to be used should be only IGF-I and GFAP positive. NIH also requires that all cells are irradiated with a dose of 5000 cGy before reinjection, to avoid the possibility of subsequent cell divisions. After irradiation, the cells must be maintained in cell culture for 24 hours.

The elimination of the plasmid in the cells before injection is ensured by the possibility of a deletion in vitro of the hygromicine for 2 to 3 weeks. (The safety of the method was guaranteed by the use of an episomal vector without the risk of integration of the DNA vector).

Gene therapy will be preceded by a lymphocyte transformation test (TTL): in vitro, the lymphocytes of patients with glioblastoma are brought into contact with their own tumor cells transfected; the increase in tritiated thymidine incorporation will allow to demonstrate the stimulation of immune cells.

The first stage is the biopsy, to harvest the cells for transfection in vitro. This step requires 4 to 6 weeks. Then, cells are frozen after verification that all cells are positive for IGF-I and GFA. In case of cellular heterogeneity after cloning, only IGF-I positive population will be frozen and preserved. The transfected cells should not produce IGF-I. The cells before injection should be irradiated. The first authorization was granted in 1994 to 12 patients [30,41].

3.3. Methodology

Primary cultures of glioma cells were etablished from biopsies of human glioblastoma patients [40]. The human glioma cell lines were transfected with plasmid vector of the "triple helix" pMT-AG TH type [26]. Clones of transfected cells, no expressing IGF-I but expressing MHC-I and B7 molecules, were selected one month after transfection. Prior to injection the transfected cells were irradiated with cGy. The first injection was performed using only cell membranes [42,43] derived from 100 000 cells, followed by two cell injection of 1 – 2 million cells, with interval of one month each (subcutaneous injection in the left arm of patients). Blood was collected before the first “membrane” vaccination, and then 3 weeks after the first and second injection. The labeling of peripheral blood lymphocytes (PBL) was performed using mouse monoclonal antibodies directed against cell surface antigens. The samples of monoclonal antibodies were used for flow cytometric analysis as follows: FITC conjugated - (a) CD45, (b) of CD4, (c) CD3, (d) CD25, (e) CD45RO, ( f) CD19 (g) CD8, (h) CD8CD11b +, (i) IgG1 control antibody, and these conjugated with PE - (a) CD14, (b) CD8, (c) CD16 + CD6, (d) CD4 (CD8), e) CD4 (CD8), (f) CD5, (g) CD8CD11b-, (h) CD8CD28, (i) IgG2 (18,27) [43].

Advertisement

4. Clinical results

The promising results were obtained in six patients at University Hospitals of Cleveland, in two patients in Bangkok and in four patients at the University Hospital of Bromberg. Admitting that the group of glioblastoma patients treated with antisense/triple helix cell injection has given the significant results, comparatively studied two cases of colon cancer and two cases of prostate cancer patients were treated, after surgery and radiotherapy with this type of “cellular therapy (Hospital of Bromberg).

Figure 1.

Antisense anti – IGF-I therapy. After in vitro transfection of tumor cells with a vector containing IGF-I cDNA in antisense orientation, the cells express IGF-I antisense RNA; the cells become negatively labeled with anti IGF-I antibodies, and positively with anti MHC-I and B7 antibodies. On the other hand, about half of these transfected cells become apoptotic. Both immune and apoptotic phenomena are related to signal transduction pathway. The injected transfected cells including apoptotic cells, together with induced in vivo APC cells, activat T lymphocytes (CTLCD8 + CD28 +). Activated CTL produce an antitumor immune response [40,44,47,61,79,80-83]. Abreviations: TAP 1,2 (transporter associated with antigen processing antigen); TK (tyrosine kinase); PI3K (phosphatidyinositol 3 kinase); PDK1 (phosphoinositide-dependent kinase 1); AKT (PKB, protein kinase B); Bcl 2 (key molecule of apoptose); GSK3 (glycogene synthetase kinase 3); GS (glycogene synthetase); MAPK (MAP kinase – mitogen activated protein kinase); PKC (protein kinaseC C).

Significant changes were observed mainly after the first cell vaccination. Phenotypic changes in peripheral blood lymphocytes were as follows: an increase in the percentage of CD8 + T cells accompanied in parallel by an increase of CD8 + CD11b- and CD8CD28 + molecules, after each of the three vaccinations. This alteration may reflect the increased activation of T cells cytotoxic blood (Figure 1.). Additionally, it was observed an increased percentage of lymphocytes positive for surface receptor of interleukin-2 (CD25). No changes were demonstrated in other CD molecules [44]. In our ongoing work (new protocol), 4 and 5 injections of IGF-I TH cells were introduced in patients with glioblastoma. After the fourth injection, the samples of the blood obtained from treated patients have shown a progressive increase in CD8 and NK cells, which highlights the effect of treatment on immune response. Also an increase of CD25 molecules was observed after the second and third injection. There was no difference before or after vaccination in the levels of CD3, CD16 + CD56, CD19, CD5, CD45 and CD14. The only side effect was a fever of 38°C noted after cell vaccinations. That probably corresponds to a cellular immune response (T cell induction). These changes may reflect the increased activation of cytotoxic T cells [36,45-47].

One glioblastoma patient who was treated at University Hospital of Cleveland, had lived 24 months from the time of diagnosis. Among other five glioblastoma patients treated in U.S.A. (University Hospital of Cleveland), two of the patients have survived 19 months. The treatment in U.S.A. has shown that the number of cell vaccines (between two and four) was not related to median survival of the patients. Another group of three patients treated in the U.S.A. have not responded so positively to therapy. These patients had an advanced disease with cerebral edema before gene therapy and were also treated with high doses of decadron or related steroids to reduce the effect of edema in CNS. Of course, this additional treatment has produced a negative effect on immune response and survival of patients. In two of the four patients with glioblastoma multiforme treated in Bromberg (NATO Science Program - USA / France / Poland / Germany), the average survival has ranged from 19 to 24 months, whereas the two patients in the control group survived 9.5 and 10 months respectively. Histopathological examination of removed glioblastoma tumors has shown that subjects had developed a necrosis around the tumor. Moreover the necrotic tissue surrounding the tumor has shown an infiltration of CD4 + and CD8 + T lymphocytes [32]. As to colon and prostate cancer patients the period of 19 months was also chosen as the end of clinical observations in all treated cancer patients. At 19 months, all these cancer patients were alive and the treatments were well tolerated. The PBL labelling results were similar to those obtained with glioblastoma patients (Figure 2. A,B,C) [48].

Figure 2.

Flow cytometric “FACS” peripheral blood lymphocyte CD marker patterns following comparative therapies in cancers of neuroectodermal origin - glioblastoma multiforme (A), entodermal origin – colon adenocarcinoma (B), and mesodermal origin – prostate adenocaarcinoma (C). CD molecules were labelled in peripheral blood lymphocytes (PBLs) obtained from prevaccinated and “vaccinated” patients. Each of the first column corresponds to data obtained before vaccinations; each second and third column corresponds to data obtained after one and two successive cellular vaccinations (IGF-I antisense/triple helix cells). Bar graphs represent the median value of the two cases. Data are expressed as percent of positive cells when compared to the isotype control. Difference in percentage of CD8+ CD11b and CD8+ CD28+ subpopulations from the relevant patients before and after vaccination was strongly significant with a range of P from 0.001 to 0.02 according to the Student’s t-test. The P value for CD8+, CD8+28+, and CD8+11b (below 0.01) is illustrated in the bar graph for statistical significance.

Advertisement

5. Discussion

The significant clinical findings were published in 2006/2012; it was demonstrated that due to the use of AS anti - IGF-I therapy following radiotherapy, the median survival of glioblastoma patients has reached 21 months. In 2010 we have reported that this survival could be explained by the immune anti-tumor response related to the increase of CD28 molecules in PBL cells analyzed after every of two "vaccinations". Moreover, this phenomenon was also observed in other tumors studied (four cases of liver cancer, colon, ovary, uterus and prostate). Phenotypic changes in peripheral blood lymphocytes (PBL) were clearly observed in all types of cancer treated with "cell therapy" - after every vaccination, an increase of CD8 + T lymphocytes, particularly of CD8+11b-, accompanied by a characteristic shift of CD8+11b+ to CD8+11b- (Figure 2.). This phenomenon was practically not significant in a group of patients treated only with "membrane" injections (as applied to two patients with glioblastoma) [47,48].

Glioblastoma and other malignancies were recently successfully treated with antisense therapy focused on TGF beta [49], using either antisense anti TGF beta expressing vector [50,51] or direct use of antisense oligodeoxynucleotides [52,53]. The use of phosphorothioate antisense oligonucleotides TGF beta2 (AP-12009, trabedersen) was initiated in patients with tumors that showed overexpression of TGF beta, such as high-grade gliomas - anaplastic astrocytoma (AA) or glioblastoma. Treatment was well tolerated. In 2007, the overall survival time was about 24 months, and in the control group, survival was 20 months. Recently, in three phase I/II studies and a randomized, active-controlled dose-finding phase IIb study, trabedersen treatment of high-grade glioma patients with recurrent or refractory tumor disease led to long-lasting tumor responses and so far promising survival data [52,54,55]. The results of clinical trials with other tumors that overexpress TGF beta have also been published recently [52,56]. Recently, the antisense approach, using also antisense oligonucleotides, targeting tumor neovascular trimer protein, laminin-411, was also proposed for clinical trial [57]. The innovation concerns a polymeric nanobioconjugate drug based on biodegradable, nontoxic, and nonimmunogenic polymalic acid as a universal delivery nanoplatform; this platform is applied for synthesis of nanomedicine drug which passes through the blood brain tumor barrier and tumor cell membrane. Other approach of antisense treatment, especially using antisense IGF-I-receptor has been developed [58-60]. It seems that this therapy, could be more efficient if the "cell vaccines" would be prepared after cell cloning for the expression of MHC-I. The different examples of antisense strategy in experimental and clinical trials of gliomas are showed in Table 1.

IGF-I Antisense vector
Clinical trial
Trojan et al. Biomed & Pharmacother 2010; 64(8):
TGF beta
& T cell therapy
Antisense oligodeoxynucleotide
Clinical trial
Dietrich et al. Curr Opin Oncol 2010; 22(6):604
Laminin-411 Antisense oligodeoxynucleotide
Clinical trial
Ding et al. Proc Natl Acad Sci USA 2010; 107(42):18143.
AKT2 Antisense oligonucleotide
Experimental therapy
Zhang et al. Oncol Rep 2010;24(1):65.
EGFR Antisense oligonucleotide
Experimental therapy
Li et al. Oncol Rep 2010; 23(6): 1585.
PED/PEA-15 (ERK1/2-
interacting protein)
Antisense oligonucleotide
Experimental therapy
Botta et al. Hum Gene Ther 2010; 21(9): 1067.
miR-21 & 5FU Antisense oligonucleotide
Experimental therapy
Ren et al. J Biomater Sci Polym Ed 2010; 21(3): 303.
miR-21 Antisense oligonucleotide
Experimental therapy
Zhou et al. Lab Invest. 2010; 90(2): 144.
EGFR Antisense oligonucleotide
Experimental therapy
Kang et al. J Biomed Mater Res A 2010; 93(2): 585.
VEGF Antisense (vector)
Experimental therapy
Yang et al. J Neurooncol 2010; Aug 26 Epub
miR-21 Antisense oligonucleotide
Experimental therapy
Zhou et al. Oncol Rep 2010; 24(1): 195.
Telomerase and tamoxifen Antisense oligonucleotide
Experimental therapy
Wang et al., Mol Med Report. 2010; 3(6): 935.
c-Met Antisense oligonucleotide
Experimental therapy
Chu et al. Oncol Rep 2010; 24(1): 189.
IGF-I Antisense vector
Clinical trial
Trojan and Anthony, Curr Signal Transd Ther. 2011; 6(3): 411.
TGF beta Antisense oligodeoxynucleotide
Clinical trial
Jashinsky et al., Curr Pharm Biotechnol. 2011; 12(12): 2203.
TGF beta Antisense oligodeoxynucleotide
Clinical trial
Hau et al., Curr Pharm Biotechnol. 2011; 12(12): 2150.
miR-10b Antisense oligonucleotide
Experimental therapy
Sun et al., Brain Res. 2011;1389: 9
micro RNA-7 Antisense oligonucleotide
Experimental therapy
Lee et al., Radiother Oncol. 2011; 101(1):171.
uPAR Antisense vector
Experimental therapy
Raghu et al., Mol Cancer. 2011; 10: 130.
IGF-I Antisense vector
Clinical trial
Trojan et al., Chem Res Pract. 2012; doi:10.1155/2012/721873
miR-221/222 Antisense oligonucleotide
Experimental therapy
Hao et al., Oncol Rep. 2012; 27(5): 1504.
miR-143 and miR-145 Antisense oligonucleotide
Experimental therapy
Koo et al., BMC Cancer. 2012; 12: 143.
miR-1275 Antisense oligonucleotide
Experimental therapy
Katsushima et al., J Biol Chem. 2012 Jun 26. [Epub]

Table 1.

Examples of experimental and clinical gene therapies of gliomas using antisense technology (selection of articles of last two years).

In the strategies of anti TGF-beta and anti IGF-I and anti IGF-IR antisense techniques, the anti-tumor immune response was designated as a primary mechanism involving growth factors inhibited by antisense technology and its signaling pathway [37,51]. As PI3K/AKT/GWK3/GS pathway is considered in antisense mechanism, it was recently demonstrated that also antisense anti-glycogen synthase (GS) cells express MHC-I molecules [44,61,62]. The IGF-I, through its binding to IGF-IR, activates the PI3K/AKT transduction cascade related to the pathway of apoptosis (IRS/PI3K/AKT/Bcl or AKT / Ca2 + or GSK3 or caspases). The end result of IGF-I AS approach involves an inhibition of the pathway elements of TK/PI3K/AKT inducing an in vivo immune response mediated by CD8 T cells and APC cells (Figure 1.) [44 ].

Advertisement

6. Conclusions

This review draws attention to the recent studies in cancer gene therapy, particularly of glioblastoma treatment using anti - gene anti IGF-I approach. Although the number of clinical trials of "antisense" type is much lower than those of pre-clinical experimental therapies, we wish to emphasize that any experimental therapy is a potential clinical trial (Table 1.). The current clinical strategies for the treatment of gliomas are usually a combination of chemotherapy and use of different types of inhibitors (imatinib, gefitinib) including antibodies (i.e. avastin), targeting growth factors and their receptors [63-66]. The new therapies are now focusing on technology of inhibitors and antigene techniques (antisense or triple helix) used alone or combined with drug treatment [67-71]. A pharmacological strategy - the use of temozolomide introduced by R. Stupp, has offered a new hope for treatment of glioblastoma. However, although median survival has reached almost two years, we are still far from victory [72,73]. The new strategies proposing to target different growth factors, especially IGF-I, TGF-beta or VEGF, their receptors and signaling pathway elements, seem to offer a promising solution [54,73-76]. We would like to underline that the research on IGF-I which has resulted in diagnostic application – the IGF-I being considered as one of the principal precancerous markers [10,12], has conducted to experiments on suppression of IGF-I expression in tumors, following directly by immuno-gene therapy of malignant tumors. Gene therapy, particularly cellular imuno-gene therapy, and cellular immunotherapy are currently among the most promising approaches for treatment of cancer diseases [66,68,77,78].

Advertisement

Acknowledgments

We thank Jean-François Nicolas (Institut Pasteur, Paris) for critical review of this manuscript, Tom Thomson Philips from Scientific Lim., London, for references related to the development of the AP-12OO9 (TGF-beta2 antisense oligonucleotides phase III clinical trial of malignant tumors). We thank Dr. Pan Yuexin (CWRU, Cleveland) for the work on the logistics of antisense IGF-I vaccines and graphic presentation of CD labeling in PBL cells, and Mrs Annabelle Trojan (CURN, Cartagena) for a revision of the text. The authors were supported by the subvention of the Society ICGT Bogota.

References

  1. 1. Trojan J, Uriel J, Deugnier MA, Gaillard J. Immunocytochemical quantitative study of alphafetoprotein in normal and neoplastic neural development. Developmental Neuroscience 1984; 6: 251-259.
  2. 2. Mollicone R, Trojan J, Oriol R. Appearance of H and B antigenes in primary sensory cells of the rat olfactory apparatus and inner ear. Developmental Brain Research 1985; 17(1): 268-279.
  3. 3. Froesch CS, Schwander J, Zapf J. Actions of insulin-like growth factors. Annual Review of Physiology 1985; 47: 443-467.
  4. 4. Daughaday WH, Rotwein P. Insulin-like growth factorsI and II. Peptide, messenger ribonucleic acid and gene structures, serum, and tissue concentrations. Endocrinology Review 1989; 10(1): 68-91.
  5. 5. Le Roith D, Bondy C, Yakar S, Liu J, Butler A. The somatomedin hypothesis. Endocrinology Review 2001; 22(1): 53-74.
  6. 6. Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors and neoplasia. Nature Review Cancer 2004; 4: 505-518.
  7. 7. Trojan J, Blossey B, Johnson T, Rudin S, Tykocinski M, Ilan J. Loss of tumorigenicity of rat glioblastoma directed by episome-based antisens cDNA transcription of insulin-like growth factor I. Proceedings of National Academy of Science USA 1992; 89: 4874-4878.
  8. 8. Werner H, Le Roith D. New concepts in regulation and function of the insulin- like growth factors: implications for understanding normal growth and neoplasia. Cellular and Molecular Life Science 2000; 57: 932-942.
  9. 9. Zumkeller W, Westphal M. The IGF/IGFBP system in CNS malignancy. Molecular Pathology 2001; 54: 227-229.
  10. 10. Zumkeller W. IGFs and IGF- binding proteins as diagnostic markers and biological modulators in brain tumours. Experimental Review of Molecular Diagnostic 2002; 2: 473-477.
  11. 11. Hajeri-Germond M, Naval J, Trojan J, Uriel J. The uptake of alphafetoprotein by C-1300 mouse neuroblastoma cells. British Journal of Cancer 1985; 51: 791-797.
  12. 12. Trojan J, Johnson TR, Rudin SD, Ilan Ju, Tykocinski ML, Ilan J. Treatment and prevention of rat glioblastoma by immunogenic C6 cells expressing antisense insulin-like growth factor I RNA. Science 1993; 259: 94-97.
  13. 13. Rubenstein JL, Nicolas JF, Jacob F. L’ARN non sens (nsARN) : un outil pour inactiver spécifiquement l’expression d’un gène donné in vivo [Nonsense RNA: a tool for specifically inhibiting the expression of a gene in vivo]. Copmtes Rendus Academie des Sciences Paris 1984; 299(8): 271-274.
  14. 14. Weintraub H, Izant JG, Harland RM. Antisense RNA as a molecular tool for genetic analysis. Trends in Genetics 1985; 1(1): 23-25.
  15. 15. Green PJ, Pines O, Inouye M. The role of antisense RNA in gene regulation. Annual Review of Biochemistry 1986; 55: 569-597.
  16. 16. Merino E, Balbas P, Puente JL, Bolivar F. Antisense overlapping open reading frames in genes from bacteria to humans. Nucleic Acids Research 1994; 22(10): 19-77.
  17. 17. Dias N, Stein CA. Basic concepts and antisense oligonucleotides mechanisms. Molecular Cancer Therapeutics 2002; 1: 347-355.
  18. 18. Galderisi U, Cascino A, Giordano A. Antisense oligonucleotides as therapeutic agents. Journal of Cell Physiology 1999; 181: 251-257.
  19. 19. Bennett CF, Butler M, Cook PD, Geary RS, Levin AA, Mehta R, Teng CL, Desmukh H, Tillman L, Hardee G. (2000). Antisense Oligonucleotides – Based Therapeutics. In: Templeton NS, Lasic DD (ed.) Gene Therapy. New York: Marcel Dekker; 2000. p305-332.
  20. 20. Tsai YJ, Hu CC, Chu CC, Imae T. Intrinsically fluorescent PAMAM dendrimer as gene carrier and nanoprobe for nucleic acids delivery: bioimaging and transfection study. Biomacromolecules 2011; 12(12): 4283-4290.
  21. 21. Scaggiante B, Morassutti C, Tolazzi G, Michelutti A, Baccarani M, Quadrifoglio E. Effect of unmodified triple helix-forming oligodeoxyribonucleotide targeted to human multidrug-resistance gene mdrl in MDR cancer cells. FEBS Letters 1994; 352: 380-384.
  22. 22. Thomas T, Faaland C, Gallo M, Thomas T. Suppression of c-myc oncogene expression by a polyamine-complexed triplex forming oligonucleotide in MCF-7 breast cancer cells. Nucleic Acids Research 1995; 23: 3594-3599.
  23. 23. Vasquez KM, Wilson IH. Triplex-directed modification of genes and gene activity. Trends in Biochemical Science 1998; 23: 4-9.
  24. 24. Dervan P. Reagents for the site-specific cleavage of megabase DNA. Nature 1992; 359:, 87-88.
  25. 25. Hélène C. Control of oncogene expression by antisense nucleic acids. European Journal of Cancer 1994; 30(A): 1721-1726.
  26. 26. Shevelev A, Burfeind P, Schulze E, Rininsland F, Johnson T, Trojan J, Chernicky C, Hélène C, Ilan Ju, Ilan J. Potential triple helix-mediated inhibition of IGF-I gene expression significantly reduces tumorigenicity of glioblastoma in an animal model. Cancer Gene Therapy 1997; 4(2): 105-112.
  27. 27. Mayfield C, Ebbinghaus S, Gee I, Jones D, Rodu B, Squibb M, Miller D. Triplex formation by the human Ha-ras promoter inhibits Spl binding and in vitro transcription. Journal of Biological Chemistry 1994; 69: 18232-18238.
  28. 28. Aggarwal B, Schwarz L, Hogan M, Rando R. Triple helix-forming oligodeoxyribonucleotides targeted to the human tumor necrosis factor (TNF) gene inhibit TNF production and block the TNF dependent growth of human glioblastoma tumor cells. Cancer Research 1996; 56: 5156-5164.
  29. 29. Rininsland F, Johnson TR, Chernicky CL, Schulze E, Burfeind P, Ilan J. Suppression of insulin-like growth factor type I receptor by a triple-helix strategy inhibits IGF-I transcription and tumorigenic potential of rat C6 glioblastoma cells. Proceedings of National Academy of Science USA 1997; 94:, 5854-5859.
  30. 30. Anthony DD, Pan Y, Wu S, Shen F, Guo Y. Ex vivo and in vivo IGF-I antisense RNA strategies for treatement of cancers in humans. Advances in Experimental Medicine and Biology 1998; 45: 27-34.
  31. 31. Trojan J, Johnson T, Rudin S, Blossey B, Kelley K, Shevelev A, Abdul-Karim F, Anthony D, Tykocinski M, Ilan Ju, Ilan J. Gene therapy of murine T eratocarcinoma: separate functions for insulin-like growth factors I and II in immunogenicity and differentiation. Proceedings of National Academy of Science USA 1994; 91: 6088-6092.
  32. 32. Wongkajornsilp A, Ouyprasertkul M, Sangruchi T, Huabprasert S, Pan, Y, Anthony DD. The analysis of peri-tumour necrosis following the subcutaneous implantation of autologous tumor cells transfected with an episome transcribing an antisense IGF-I RNA in a glioblastoma multiforme subject. J Med Assoc Thai 2001; 4(3): 740-747.
  33. 33. Brooks WH, Latta RB, Mahaley MS. Immunobiology of primary intracranial tumors. Journal of Neurosurgery 1981; 54: 331-337.
  34. 34. Saji M, Moriarty J, Ban T, Singer D, Kohn L. Major Histocompatibility Complex class I gene expression in rat thyroid cells is regulated by hormones, methimazoleand iodide as well as interferon. Journal of Clinical Endocrinology and Metabolism 1992; 75(3): 871-878.
  35. 35. Blanchet O, Bourge JF, Zinszner H, Israel A, Kourilsky P, Dausset J, Degos L, Paul P. Altered binding of regulatory factors to HLA class I enhancer sequence in human tumor cell lines lacking class I antigen expression. Proceedings of National Academy of Science USA 1992; 89(8): 3488-3492.
  36. 36. Schwartz RH. Costimulation of T Iymphocytes: the role ofCD28, CTLA-4 and B7/BBI in interleukin-2 production and immunotherapy. Cell 1992; 71: 1065- 1068.
  37. 37. Trojan J, Duc HT, Upegui-Gonzalez L, Hor F, Guo Y, Anthony DD, Ilan J. Presence of MHCI and B-7 molecules in rat and human glioma cells expressing antisense IGF-I mRNA. Neuroscience Letters 1996; 212: 9-12.
  38. 38. Lafarge-Frayssinet C, Duc HT, Sarasin A, Frayssinet C, Anthony D, Guo Y, Trojan J. Antisense IGF-I transfer into a rat hepatoma cell line inhibits tumorigenesis by modulatig MHC-I. Cancer Gene Therapy 1997; 4(5): 276-285.
  39. 39. Upegui-Gonzalez LC, Duc HT, Buisson Y, Arborio M, Lafarge-Frayssinet C, Jasmin C, Guo Y, Trojan J. Use of antisense strategy in the treatment of the hepatocarcinoma. Advances in Experimental Medicine and Biology 1998; 451: 35-42.
  40. 40. Ly A, Duc HT, Kalamarides M, Trojan LA, Pan Y, Shevelev A, François J-C, Noël T, Kane A, Henin D, Anthony DD, Trojan J. Human glioma cells transformed by IGF-I triple-helix technology show immune and apoptotic characteristics determining cell selection for gene therapy of glioblastoma. Journal of Clinical Pathology (Molecular Pathology) 2001; 54(4): 230-239.
  41. 41. Ilan J., editor. Clinical trial: Gene therapy for human brain tumors using episome based antisense cDNA transcription of Insulin like Growth Factor I. Proposal for a phase one gene therapy clilnical study. NIH no 1602. Bethesda, Maryland: NIH; 1993.
  42. 42. Matlib MA, Kihara M, Farrell C, Dage RC. The Na+-Ca2+ exchange system in vascular smooth muscle cell membrane vesicles isolated from cultured cells and from tissue is similar. Biochimical and Biophysical Acta 1988; 939(1): 173-177.
  43. 43. Trojan LA, Ly A, Kopinski P, Ardourel M-Y, Dufour T, Duc HT, Kasprzak H, Cloix J-F, Wei MX, Chyczewski L, Pan Y, Chatel M, Anthony DD, Trojan J. Antisense and triple helix anti IGF-I tumours vaccines - gene therapy of gliomas. International Journal of Cancer Prevention 2007; 2(4): 227-243.
  44. 44. Trojan J, Cloix J-F, Ardourel M-Y, Chatel M, Anthony D. IGF-I biology and targeting in malignant glioma. Neuroscience 2007; 145(3): 795-812.
  45. 45. Chen L, Ashe S, Brady WA, Hellstrom KE, Ledbetter IA, Mc Growan P, Linsley PS. Costimulation of anti-tumor immunity by the B7 counter receptor for the T lymphocyte molecules CD28 and CTLA-4. Cell 1992; 71: 1093-1102.
  46. 46. Matthew L, Saiter B, Bhardwag N. Dendritic cells acquire antigen from apoptotic cells and induce class I restricted CTL. Nature 1998; 392: 86-89.
  47. 47. Trojan J, Ly A, Wei MX, Kopinski P, Ardourel M-Y, Pan Y, Trojan LA, Dufour D, Shevelev A, Andres C, Chatel M, Kasprzak H, Anthony DD, Duc HT. Antisense anti-IGF-I cellular therapy of malignant tumours: immune response in cancer patients. Biomedicine & Pharmacotherapy 2010; 64(8): 576-578.
  48. 48. Trojan J, Pan YX, Wei MX, Ly A, Shevelev A, Bierwagen M, Ardourel M-Y, Trojan LA, Alvarez A, Andres C, Noguera MC, Briceño I, Aristizabal BH, Kasprzak H, Duc HT, Anthony DD. Methodology for anti - gene anti - IGF-I therapy of malignant tumours. Chemotherapy Research and Practice 2012; doi: 10.1155/2012/721873.
  49. 49. Kaminska B, Wesolowska A, Danilkiewicz, M. TGF beta signalling and its role in tumour pathogenesis. Acta Biochimica Polonica 2005; 52: 329-337.
  50. 50. Fakhrai H, Dorigo O, Shawler DL, Lin H, Mercola D, Black KL, Royston Y, Sobol RE. Eradication of established intracranial rat gliomas by transforming growth factor beta antisense gene therapy. Proceedings of National Academy of Science USA 1996; 93(7): 2909-2914.
  51. 51. Fakhrai H, Mantil JC, Liu L, Nicholson GL, Murphy-Satter CS, Ruppert J. Phase I clinical trial of a TGF-beta antisense-modified tumor cell vaccine in patients with advanced glioma. Cancer Gene Therapy 2006; 13(12): 1052-1060.
  52. 52. Schlingensiepen KH, Schlingensiepen R, Steinbrecher A, Hau P, Bogdahn U, Fischer-Blass B, Jachimczak P. Targeted tumor therapy with the TGF-beta2 antisense compound AP 12009. Cytokine Growth Factor Review 2006; 17: 129-139.
  53. 53. Schlingensiepen KH, Fischer-Blass B, Schmaus S, Ludwig S. Antisense therapeutics for tumor treatment: the TGF-beta2 inhibitor AP 12009 in clinical development against malignant tumors, Recent Results Cancer Research 2008; 177: 137-150.
  54. 54. Hau P, Jachimczak P, Bogdahn U. Treatment of malignant gliomas with TGF-beta2 antisense olgonucleotides. Expert Review Anticancer Therapy 2009; 9(11): 1663-1674.
  55. 55. Hau P, Jachimczak P, Schlaier J, Bogdahn U. TGF-β2 signaling in high-grade gliomas. Current Pharmaceutical Biotechnology 2011; 12(12): 2150-2157.
  56. 56. Schlingensiepen KH, Jaschinski F, Lang SA, Moser C, Geissler EK, Schlitt HJ, Kielmanowicz M, Schneider A. Transforming growth factor-beta 2 gene silencing with trabedersen (AP 12009) in pancreatic cancer. Cancer Science 2011; 102(6): 1193-1200.
  57. 57. Ding H, Inoue S, Ljubimov AV, Patil R, Portilla-Arias J, Hu J, Konda B, Wawrowsky KA, Fujita M, Karabalin N, Sasaki T, Black KL, Holler E, Ljubimova JY. Inhibition of brain tumor growth by intravenous poly (β-L-malic acid) nanobioconjugate with pH-dependent drug release [corrected]. Proceedings of National Academy of Science USA. 2010; 107(42): 18143-18148.
  58. 58. Resnicoff M, Li W, Basak S, Herlyn D, Baserga R, Rubin R. Inhibition of rat C6 glioblastoma tumor growth by expression of insulin-like growth factor I receptor antisense mRNA. Cancer Immunology and Immunotherapy 1996; 42: 64-68.
  59. 59. Andrews DW, Resnicoff M, Flanders AE, Kenyon L, Curtis M, Merli G, Baserga R, Iliakis G, Aiken RD. Results of a pilot study involving the use of an antisense oligodeoxynucleotide directed against the insulin like growth factor type I receptor in malignant astrocytomas. Journal of Clinical Oncology 2001; 19: 2189-2200.
  60. 60. Baserga R. The insulin-like growth factor-I receptor as a target for cancer therapy. Expert Opinion on Therapeutic Targets 2005; 9: 753-768.
  61. 61. Premkumar DR, Arnold B, Jane EP, Pollack IF. Synergistic interaction between 17 AAG and phosphatidylinositol 3-kinase inhibition in human malignant glioma cells. Molecular Carcinogene 2006; 45: 47-59.
  62. 62. Ardourel M-Y, Blin M, Moret J-L, Dufour T, Duc HT, Hevor T, Trojan J, Cloix J-F. A new putative target for antisense gene therapy of glioma: glycogen synthetase. Cancer Biology and Therapy 2007; 6(5): 719-723.
  63. 63. Stupp R, Hegi ME, van den Bent MJ, Mason WP, Weller M, Mirimanoff RO, Cairncross JG. Changing paradigms--an update on the multidisciplinary management of malignant glioma. Oncologist 2006; 11: 165-180.
  64. 64. Reardon DA, Quinn JA, Vredenburgh JJ, Gururangan S, Friedman AH, Desjardins A, Sathornsumetee S, Herndon JE 2nd, Dowell JM, McLendon RE, Provenzale JM, Sampson JH, Smith RP, Swaisland AJ, Ochs JS, Lyons P, Tourt-Uhlig S, Bigner DD, Friedman HS, Rich JN. Phase 1 trial of gefitinib plus sirolimus in adults with recurrent malignant glioma. Clinical Cancer Research 2006; 12: 860-868.
  65. 65. Wen PY, Yung WK, Lamborn KR, Dahia PL, Wang Y, Peng B, Abrey LE, Raizer J, Cloughesy TF, Fink K, Gilbert M, Chang S, Junck L, Schiff D, Lieberman F, Fine HA, Prados MD. Phase I/II study of imatinib mesylate for recurrent malignant gliomas: North American Brain Tumor Consortium Study 99-08. Clinical Cancer Research 2006; 12: 4899-4907.
  66. 66. Cavazzana-Calvo M, Hacein-Bey-Abina S, Fischer A: Ten years of gene therapy: thoughts and perspectives: Medecine Science (Paris) 2010; 26(2): 115-118.
  67. 67. Giovannangeli C, Hélène C. Progress in developments of triplex-based strategies. Antisense Nucleic Acid Drug Development 1997; 7: 413-421.
  68. 68. Dietrich PY, Dutoit V, Tran Thang NN, Walker PR. (2010). T cell immunotherapy for malignant glioma: toward a combined approach. Current Opinion in Oncology 2010; 22(6): 604-610.
  69. 69. Bodera P. Immunostimulatory oligonucleotides. Recent Pathology Inflammation Allergy Drug Discovery 2011; 5(1): 87-93.
  70. 70. Piwecka M, Rolle K, Wyszko E, Żukiel R, Nowak S, Barciszewska MZ, Barciszewski J. Nucleic acid-based technologies in therapy of malignant gliomas. Current Pharmaceutical Biotechnology 2011; 12(11): 1805-1822.
  71. 71. Catuogno S, Esposito CL, Quintavalle C, Condorelli G, de Franciscis V, Cerchia L. Nucleic acids in human glioma treatment: innovative approaches and recent results. Journal of Signal Transduction 2012; 2012:735135. Epub 2012 May 21.
  72. 72. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JE, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC, Stupp R. MGMT gene silencing and benefit from temozolomide in glioblastoma. New England Journal of Medicine 2005; 352: 997-1003.
  73. 73. Gorlia T, van den Bent MJ, Hegi ME, Mirimanoff RO, Weller M, Cairncross JG., Eisenhauer E, Belanger K, Brandes AA, Allgeier A, Lacombe D, Stupp R. Nomograms for predicting survival of patients with newly diagnosed glioblastoma: prognostic factor analysis of EORTC and NCIC trial 26981 22981/CE.3. Lancet Oncology 2008; 9(1): 29-38.
  74. 74. Ellouk-Achard S, Djenabi S, De Oliveira GA, Dessay G, Duc HT, Zoar M, Trojan J, Claude JR, Sarasin A, Lafarge-Frayssinet C. Induction of apoptosis in rat hepatoma cells by expression of IGF-I antisense cDNA. Journal of Hepatology 1998; 29: 807-818.
  75. 75. Goudar RK, Shi Q, Hjelmeland MD, Keir ST, McLendon RE, Wikstrand CJ, Reese ED, Conrad CA, Traxler P, Lane HA, Reardon DA, Cavenee WK, Wan XF, Bigner DD, Friedman HS, Rich JN. Combination therapy of inhibitors of epidermal growth factor receptor/vascular endothelial growth factor receptor 2 (AEE788) and the mammalian target of rapamycin (RAD001) offers improved glioblastoma tumor growth inhibition. Molecular Cancer Therapy 2005; 4: 101-112.
  76. 76. Pan Q, Luo X, Chegini N. Blocking neuropilin-1 function has an additive effect with anti-VEGF to inhibit tumor growth. Cancer Cell 2007; 11(1): 53-67.
  77. 77. Lemoine FM, Cherai M, Giverne C, Dimitri D, Rosenzwajg M, Trebeden-Negre H, Chaput N, Barro B, Thioun N, Gattegnio B, Selles F, Six A, Azar N, Lotz JP, Buzyn A, Sibony M, Delcourt A, Boyer O, Herson S, Klatzmann D, Lacave R. Massive expansion of regulatory T-cells following interleukin 2 treatment during a phase I-II dendritic cell-based immunotherapy of metastatic renal cancer. International Journal of Oncology 2009; 35(3): 569-581.
  78. 78. Iwami K, Natsume A, Wakabayashi T. Gene therapy for high-grade glioma. Neurologia Medico-Chirurgica (Tokyo) 2010; 50(9): 727-736.
  79. 79. Fonteneau JF, Larsson M, Bhardwaj N. Interactions between dead cells and dendritic in the induction of antiviral CTL responses. Current Opinion in Immunology 2002: 14: 471-477.
  80. 80. Lebedeva IV, Stein CA. Antisense Down Regulation of the Apoptosis – Related bcl-2 and bcl-xl Proteins: a New Approach to Cancer Therapy. In: Lattime EC, Gerson SL (ed.) Gene Therapy of Cancer. New York; Academic Press: 2002. p315-330.
  81. 81. Patel S, Doble B, Woodgett JR. Glycogen synthase kinase-3 in insulin and Wnt signalling: a double-edged sword?. Biochemical Society Transversal 2004; 32: 803-808.
  82. 82. Beckner ME, Gobbel GT, Abounader R, Burovic F, Agostino NR, Laterra J, Pollack IF. Glycolytic glioma cells with active glycogen synthase are sensitive to PTEN and inhibitors of PI3K and gluconeogenesis. Laboratory Investigation 2005; 85: 1457-1470.
  83. 83. Lo HW. Targeting Ras-RAF-ERK and its interactive pathways as a novel therapy for malignant gliomas. Current Cancer Drug Targets 2010; 10(8): 840–848.

Written By

Jerzy Trojan and Ignacio Briceno

Submitted: 08 August 2012 Published: 27 February 2013