Open access peer-reviewed chapter

Oncoproteins Targeting: Antibodies, Antisense, Triple-helix. Case of Anti IGF-I Cancer Immunogene Therapy

Written By

Trojan Jerzy

Submitted: 31 July 2018 Reviewed: 15 November 2018 Published: 15 March 2019

DOI: 10.5772/intechopen.82548

From the Edited Volume

Antisense Therapy

Edited by Shashwat Sharad and Suman Kapur

Chapter metrics overview

1,016 Chapter Downloads

View Full Metrics

Abstract

AFP and IGF-I oncoproteins were introduced as biomarkers for cancer diagnosis and targeted in cancer therapy on protein level, but also on transcription and translation levels. The protein level was targeted using an injection of antibodies or radiolabeled proteins. The transcription and translation levels were targeted by triple helix and antisense technologies, respectively. AFP was especially useful for diagnosis and therapy of liver cancer, IGF-I was applied in diagnosis and therapy of colon, prostate, liver, uterus, ovary and brain tumors. The most spectacular results were obtained with IGF-I anti-gene strategy. IGF-I antisense (AS)/triple helix (TH) gene therapy was successfully introduced in clinical trial in the USA and Europe. When using IGF-I anti-gene therapy, cancer cells provided from biopsies were transfected in vitro with IGF-I AS, IGF-I TH expression vectors. A decrease in IGF-I gene expression of 80 and 60% was demonstrated when using TH and AS technologies, respectively. These transfected cells expressing MHC-I molecules, while injected in vivo, induced immune antitumor response mediated by CD8 lymphocytes. The median survival of treated glioblastoma patients was 21–22 months. IGF-I AS/TH immunogene therapy constitutes one of the most promising approaches in cancer therapy, and more specifically when it comes to glioblastoma treatment.

Keywords

  • cancer
  • glioma
  • alpha-fetoprotein
  • IGF-I
  • diagnostic
  • antibodies
  • antisense
  • triple helix
  • immunotherapy

1. Introduction

Oncoproteins like alpha-fetoprotein (AFP), serum albumin (AS), and growth factors such as GH, IGF, TGF-beta, and EGF are present in embryo-fetal tissues and reappear in neoplastic developing tissues, including the central nervous system; this concerns especially AFP [1, 2, 3, 4, 5, 6, 7, 8] and IGF-I [9, 10, 11, 12, 13, 14, 15, 16, 17]. AFP is present in both neural and glial developing and cancerous cells, whereas IGF-I is only present in glial developing and tumoral cells [10, 18]. This striking difference has helped us orientate a strategy to manage the most malignant brain tumor expressing IGF-I gene—glioblastoma.

IGF-I plays an important role in growth as a mediator of growth hormone [19, 20, 21]. Blocking IGF-I synthesis induces apoptotic and immunogenic phenomena [12, 22]. Both phenomena, apoptosis and immunogenicity, related to arrest of IGF-I expression in neoplastic glial cells, were used to prepare antitumor cell vaccines for therapy of brain malignant tumor—glioblastoma [12, 23].

An efficient strategy targeting IGF-I was established by construction of vectors stopping the synthesis of this oncoprotein on translation and transcription levels: vectors expressing either IGF-I antisense RNA or IGF-I RNA forming RNA-DNA triple helix, respectively. The glioma cells transfected with these vectors, when injected in vivo in animals bearing tumors like glioma or teratocarcinoma or applied in clinical treatment of glioblastoma patients, induced an immune antitumor effect (CD8+) accompanied by increase of the median survival of patients (successful clinical results obtained in the USA, E.U., China) [22, 23].

Advertisement

2. Oncoproteins in diagnostic: AFP and IGF-I

2.1 AFP

AFP is present in normal and neoplastic developing tissues [1, 24, 25, 26]. The presence of AFP seems to be related to the stage of cell and tissue differentiation. AFP is absent from either undifferentiated or fully differentiated cells [24].

The localization of AFP was compared with that of another oncoprotein—serum albumin, SA. SA-mRNA gave a strong signal in differentiating structures as well as in undifferentiated cell clusters. AFP-mRNA was observed only in differentiating structures; this observation was especially useful in the clinical diagnostic of hepatocarcinoma [4, 27].

During an experiment with teratocarcinoma-bearing mice injected intraperitoneally with J-125 radiolabeled SA and AFP, significant accumulations of both SA and AFP were demonstrated in the tumors, SA being about 3-fold higher than that of AFP after normalization to quantity of uptake in liver. In the case of comparatively studied neuroblastoma presenting only neuroblastic components (different from teratocarcinoma containing both neuroectoblastic and neuroblastic elements), the accumulation of radiolabeled SA and AFP showed relationship 1:1. External in vivo photo scanning confirmed this relationship of accumulated radiolabeled proteins in both studied tumors; the last observations were useful for differential diagnosis of tumors [4, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36].

AFP may be used to advantage in radio tracing experiments, since this isologous protein is not expected to induce hypersensitivity reactions. On the other hand, and contrary to SA, the extremely low serum levels of AFP in adult individuals should minimize effects due to competition with endogenous protein. This makes AFP a good candidate for tumor biomarker by imaging techniques. The diagnosis and therapies of CNS tumors including neuroblastoma are always a subject of discussion [36, 37, 38, 39].

2.2 IGF-I

Another oncodevelopmental antigen, an insulin like-growth factor, IGF-I [20, 40, 41, 42, 43], is present in glioma cells but absent in neuroblastoma cells [18]; neuroblastic cells express IGF-II [27]. These observations permitted to study separately, using IGF-I and IGF-II as the oncoprotein markers, glial and neural tumors [13, 15, 20, 40, 41, 42, 44, 45, 46].

Comparative studies of AFP, IGF-I, IGF-II presence in neoplastic cells [3, 4, 18] have demonstrated that IGF-I constitutes an essential target for genetic testing. IGF-I, similarly to AFP, is involved in tissue development and differentiation, especially in the development of the nervous system [9, 15, 19, 20, 47, 48, 49, 50, 51]. According to Baserga [43], IGF-I is one of the most important growth factors related to normal and neoplastic differentiation [50, 52, 53, 54].

The elements of IGF-I related transduction pathway (IRS/PI3K-PKC/PDK1/AKT-Bcl2/GSK3/GS) [55, 56] were also considered as targets for diagnostic [9, 55, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66]. The relationship between IGF-I and IGF binding proteins are being introduced in clinical diagnostics as one of the indicators of precancerous development [67].

IGF-I becomes useful in molecular diagnostic of neonatal CNS malformations and tumors [5, 9, 13, 39, 68, 69]. Diagnosis and treatment should logically be related, at first using IGF-I gene testing for diagnosis [13, 21, 70], and then targeting IGF-I gene through special therapy, such as cancer gene therapy, especially therapy of gliomas [11, 22, 71, 72, 73].

Advertisement

3. IGF-I and anti-gene immunotherapy

3.1 Protein, translation and transcription levels

To target an oncoprotein directly on protein level, the strategy of antibodies was explored. Cancers treated by use of antibodies was in general not efficient.

The treatment of any cancer, especially hepatocarcinoma, demands a permanent perfusion, per vena porta, of anti AFP antibodies. The arrest of perfusion has produced the reappearance of cancer. Similar observation has been made when using antibodies against growth factors.

The only possibility was to stop the synthesis of the oncoprotein on the translation or on the transcription level of the concerned gene, and directly in the cancer cells. This hypothesis and our knowledge of chemistry was an epistemological problem, pointed out by Mosquera “how to integrate the knowledge of chemistry with technique” [74].

As to glioma malignant tumor, glioblastoma (the mortality remains close to 100%), new or proposed therapies are based generally either on immune treatment or on immuno-gene strategies [75, 76]. In order to define new therapies, the different techniques for inhibitors [9], and the anti-gene strategy (either antisense, AS, or triple helix, TH, approaches) were investigated [61] (Figure 1).

Figure 1.

Schema of technologies suppressing the expression of oncoproteins, including growth factors, GF: antibodies, antisense and triple helix. In antibodies technology, the GF expression is directly blocked by its ligand—antibody. In antisense technology, the end result is the inhibition of GF mRNA (sense RNA) activity by binding to the antisense RNA. In triple helix technology, the oligopurine third strand (23 bp) forms RNA-DNA triple helix with GF gene.

The AS technology [77, 78] has permitted us to establish new and successful gene therapy strategies targeting glioma’s growth factors [11, 79] and have now been introduced into clinical trials. Other recently introduced technologies include those of triple helix, TH [80, 81, 82, 83], as well as potentially useful siRNA [84, 85] and miRNA (microRNA) [82, 83, 86, 87]. The role of 21–23 mer double-stranded RNA (siRNA) in the silencing of genes is strongly similar to that of the TH DNA mechanism, which also involves 23 mer RNA [81]. Whether or not siRNA technology or miRNA knockdown will supplant the AS and TH oligodeoxynucleotide approaches remains in question at this time [83, 85, 87, 88, 89, 90]. AS methodology is currently being standardized to be largely used in clinical trials [18, 89].

As to TH strategy, the oligonucleotides are targeted to double stranded DNA containing polypurine-polypyrimidine sequences that readily form triple helixes. The studies of triple helix strategy have shown that an RNA strand containing a 23-nucleotide (nt) oligopurine sequence [80, 86, 87] may be capable of forming triple helix structures with an oligopurine-oligopyrimidine sequence of the IGF-I gene as well in cultured rat C6 glioma as in rat CNS-1 glioma, and in mouse PCC-4 cells [86]. Although we cannot exclude other mechanisms, triple helix formation remains the most plausible possibility for the inhibition of IGF-I gene expression [86]. The arrest of IGF-I synthesis suggests that the RNA strand, which forms the triple helix, has inhibited gene transcription in glioma cells.

3.2 Antisense and triple helix experimental studies

In our experimental studies, we have shown that in IGF-I “antisense” and “triple helix” transfected CNS-1 rat glioma cells, PCC-4 mouse embryonal carcinoma cells, and in primary human glioma cells, changes in immunogenic properties and apoptosis occur. The induction of IGF-I triple helix forming structure, similarly to IGF-I antisense approach, was followed by enhanced expression of MHC-I and B-7 [5, 18, 90, 91, 92, 93, 94, 95] and loss of in vivo tumorigenicity. An extensive lymphocytic CD8 positive infiltration 4–5 days after injection of AS transfected glioma, teratocarcinoma and hepatoma cells into the respective animal bearing tumors was demonstrated [11, 27, 96]. These properties were used for selection of human glioma cells that were used in IGF-I antisense/triple helix immunogene therapy [90] (Figure 2).

Figure 2.

Schema of cancer immunogene therapy. The cells isolated from tumor biopsy are growing in tissue culture. The established cell line is transfected by vector of anti-gene type (antisense and triple helix IGF-I). The transfected cells, and originated apoptotic cells, are injected in proportion 50–50 (“vaccine”) in the cancer patients. The presence of MHC-I and B7 molecules present in transfected tumor cells induce an immune response mediated by T lymphocytes. Moreover, APC cells also participate as shown in the immunogene mechanism. The activated T lymphocytes destroy as well the vaccine (transfected tumor cells) as a tumor.

In this context, antigenic peptides presented by class I MHC molecules were necessary but, in general, not sufficient to stimulate T cell response. In the absence of B-7 molecule, MHC-peptide complexes could selectively inactivate T-cells [97]. (Although “triple helix” cells as compared to “antisense” cells show slightly higher expression of MHC-I and B7 there are no qualitative immunogenic and apoptotic differences between IGF-I “antisense” and “triple helix” approaches).

The absence of IGF-I synthesis in AS and TH transfected cells, could lead to a higher level of IGF-I receptor and hence to greater tyrosine kinase content [98]. There is a relation between the signal transduction pathway of tyrosine kinase and induction of B-7 molecules: enhancement in B-7 co-stimulation through a cAMP mechanism linked to tyrosine kinase of the CD 28 receptor has been previously reported [99]. Similar signaling through the tyrosine kinase activity of the IGF-I receptor shows that: tyrosine kinase activates IRS-1 (Insulin receptor substrate-1), and then IRS-1 activates PI3K (phosphatidylinositol 3 kinase) [100, 101]. This mechanism could be considered in the cytokine induced B7–1 expression demonstrated in fetal human microglia in culture [102].

In the AS and TH transfected cells, IGF-I-R activated by its ligand plays a very protective role in programmed cell death, and that this protection is even more striking in vivo than in vitro [103]. Apoptosis could play a specific role in our strategies; the phenotypic modifications due to apoptosis may explain the recognition of the transfected cells by the immune system like tumor-specific immunity mediated by CD8+ T [11, 98]. Apoptotic cells, in the context of MHC-I are recognized by dendritic cells activating lymphocytes T-CD8 [104, 105]. B-7 molecules can be included in this mechanism, because both MHC-I and B-7 molecules are necessary for T cell activation [16, 27, 36, 91, 106, 107, 108].

While working on mouse hepatoma cells transfected by the IGF-I triple helix approach, we have observed a decrease of cytokines such as Il-10, which is a strong immunosuppressor [109], and TNF-alpha, which can act as a factor stimulating tumor growth [110, 111]. Moreover, we have found increased levels of TAP 1 and 2 in these cells. The relationship between the immune process, related to the MHC-I or HLA system [112, 113], and the apoptotic process is under study.

3.3 Anti-gene clinical therapy

In the clinical trial using anti-gene IGF-I therapy after subcutaneous vaccination, the subject developed peri-tumor necrosis; the tissue section bordering the necrotic tumor tissue showed infiltration of lymphocytes consisting of both CD4 and CD8 T cells [114]. Other results concerning peripheral blood lymphocytes (PBL) of glioblastoma patients treated by IGF-I triple helix immuno-gene therapy show eminent changes in CD8 T cells, especially after second injection. There was also an increase in the percentage of CD8 with characteristics switching from CD8+ CD11b+ to CD8+ CD11b− phenotype, an alteration that may reflect the enhanced activation of T cytotoxic cells in blood. Additionally, as far as prognosis of glioblastoma patients is concerned, the patients have survived between 18 and 24 months after the first diagnosis. Generally, patients with diagnosed glioblastoma multiforme, undergoing surgeries and radiotherapy, survive up to 14 months [22, 113].

Our phase I human cancer clinical trial based on the anti-gene, antisense approach, is in progress in the U.S.A, Thailand, Poland and China [107, 114, 115] and is in parallel with previously established approaches of glioblastoma treatment established by either Culver [116] or by Baserga [65]. Following the same approach, different antisense strategies to treat glioma tumors have been recently investigated [26, 31, 117, 118], especially in relation to glycogen metabolism [119] in glioma cells. (Malignant astrocytes present a high level of glycogen [55]). The therapy of gliomas using strategies of growth factors inhibition is in permanent progress. The inhibition of the IGF-I receptor using AS technology, and its signaling pathway has opened a door for experimental and clinical research in various tumors [9, 14, 15, 42, 62, 120, 121].

Advertisement

4. Discussion

The use of anti-gene therapy was also effective in another AS approach, targeted toward the molecule, TGF-beta [79]. TGF-beta2 plays a role in tumor progression by regulating key mechanisms including proliferation, metastasis, and angiogenesis. The approach of AS TGF-beta using an AS oligodeoxynucleotide—compound AP 12009, has given satisfactory results [122, 123, 124]. AP-12009 treatment was well tolerated and tumor response has been observed [123]. Two patients experienced long-lasting complete tumor remission [124]. In another clinical AS TGF-beta study, a phase I clinical trial in grade IV astrocytoma (GBM) was performed using autologous tumor cells modified by a AS TGF-beta2 vector [79]. There were indications of humoral and cellular immunity induced by the vaccine [57, 79].

As far as AS TGF-beta therapy of GBM is concerned, the treatment of patients with recurrent or refractory malignant (high-grade) glioma, WHO grade III or IV has been shown to produce results, similar to results obtained with anti IGF-I treatment. The role of peripheral blood mononuclear cells in the immune antitumor response, and prolonged survival in both anti TGF beta and anti IGF-I approaches was comparatively examined. In the case of TGF-beta, it is also important to mention, that although the first successful clinical results were published in 2006–2008, the solid experimental data, using AS technology were obtained in 1994/95 [79], which means, that long periods of research on the mechanism of AS TGF-beta and AS IGF I are required before achieving significant clinical results, confirming the usefulness of gene therapy in cancer treatment [90, 122, 123, 125, 126, 127, 128, 129, 130].

In AS IGF-I or AS TGF-beta approaches, immune antitumor response, mediated by TCD8 and APC cells, was signaled as a principal mechanism of AS technology inhibiting growth factors and their signaling pathway [12, 79]. These cells being involved in HS (heat shock) protein mechanism, the inhibition of HS was introduced in clinical trials as a new direction for cancer therapy [131].

More recent clinical successful strategies of gliomas treatment, generally as a combination therapy using different types of inhibitors (i.e., imatinib, gefitinib) including antibodies (i.e., avastin) targeting growth factors and their receptors [132, 133, 134, 135, 136], are now focusing on anti-gene strategy, especially on antisense or triple helix technologies used alone or combined also with pharmacological treatment. As far as antisense and triple helix strategies are compared, the triple helix blockade of growth factor has given better results in vitro and in vivo. But combining both strategies, the final result is most effective—stopping the expression of the oncoproteins on translation (AS) and transcription (TH) levels, respectively.

Advertisement

5. Conclusion

Among the new strategies in the efforts to successfully treat GBM, the use of AS approach targeting IGF-I, TGF-beta or VEGF, their receptors and their downstream transduction signaling elements [9, 137], appears to offer a promising solution. The final result of the signal transduction pathway element inhibition is an immune response mediated in vivo by lymphocytes T CD8 and APC cells. Using cancer immunogene therapy of anti-gene anti IGF-I approach, the median survival of treated glioblastoma patients has reached 22–24 months. But the near future in treating this group of disorders belongs to a combination of treatment: classical surgery, radiotherapy with immunotherapy, pharmacologic therapy, growth factor inhibitors, and the use of the antisense/triple helix gene blockade approach targeting signal transduction pathway elements of cancer processes [16, 22, 23, 36, 66, 90, 125, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147].

Advertisement

Conflict of interest

No conflict of interest.

References

  1. 1. Abelev GJ. Alpha-fetoprotein in ontogenesis and its association with malignant tumors. Advances in Cancer Research. 1971;14:295-299
  2. 2. 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
  3. 3. Hajeri-Germond M, Naval J, Trojan J, et al. The uptake of alphafetoprotein by C-1300 mouse neuroblastoma cells. British Journal of Cancer. 1985;51:791-797
  4. 4. Trojan J, Naval X, Johnson T, et al. Expression of serum albumin and of alphafetoprotein in murine normal and neoplastic primitive embryonic structures of teratocarcinoma. Molecular Reproduction and Development. 1995;42(4):369-378
  5. 5. Harding BN, Golden JA. Developmental Neuropathology. Basel, Switzerland: International Society of Neuropathology; 2004
  6. 6. Calaminus G, Bamberg M, Harms D, et al. AFP/β-HCG secreting CNS germ cell tumors: Long-term outcome with respect to initial symptoms and primary tumor resection. Results of the Cooperative Trial MAKEI 89. Neuropediatrics. 2005;36(2):71-77
  7. 7. Kim A, Ji L, Balmaceda C, et al. The prognostic value of tumor markers in newly diagnosed patients with primary central nervous system germ cell tumors. Pediatric Blood & Cancer. 2008;51(6):768-773
  8. 8. Kawaguchi T, Kumabe T, Kanamori M, et al. Logarithmic decrease of serum alpha-fetoprotein or human chorionic gonadotropin in response to chemotherapy can distinguish a subgroup with better prognosis among highly malignant intracranial non-germinomatous germ cell tumors. Journal of Neurooncology. 2011;104(3):779-787
  9. 9. Trojan J, Cloix J-F, Ardourel M, et al. IGF-I biology and targeting in malignant glioma. Neuroscience. 2007;145(3):795-811
  10. 10. Kiess W, Lee L, Graham DE, et al. Rat C6 glial cells synthesize insulin-like growth factor I (IGF-I) and express IGF-I re-ceptors and IGF-II/mannose 6-phosphate re-ceptors. Endocrinology. 1989;124:1727-1736
  11. 11. Trojan J, Johnson T, Rudin S, et al. Treatment and prevention of rat glioblastoma by immugenic C6 cells expressing antisense insulin-like growth factor I RNA. Science. 1993;259:94-97
  12. 12. Ly A, Duc HT, Kalamarides M, et al. Human glioma cells transformed by IGF-I tri-ple-helix technology show immune and apoptotic characteristics determining cell selection for gene therapy of glioblastoma. Journal of Clinical Pathology (Molecular Pathology). 2001;54:230-239
  13. 13. Zumkeller W. IGFs and IGF-binding proteins as diagnostic markers and biological modulators in brain tumors. Expert Review of Molecular Diagnostics. 2002;2:473-477
  14. 14. Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors and neoplasia. Nature Reviews. Cancer. 2004;4:505-518. DOI: 10.1038/nrc1387
  15. 15. Baserga R. The insulin-like growth factor-I receptor as a target for cancer therapy. Expert Opinion on Therapeutic Targets. 2005;9:753-768
  16. 16. Pan Y, Trojan J, Guo Y, et al. Rescue of MHC-1 antigen processing machinery by down-regulation in expression of IGF-I in human glioblastoma cells. PLoS One. 2013;8(3):e58428. DOI: 10.1371/0058428
  17. 17. Trojan A, Jay LM, Briceño I, et al. IGF-I, IGF-I gene and diagnostic. In: Trojan J, editor. Cancer Immunogene Therapy. Anti-Gene Anti IGF-I Approach. Case of Glioblastoma. Saarbrucken, Germany: Lambert Academic Publishers; 2017. pp. 7-28. ISBN-13: 978-3-330-06345-7; ISBN-10: 3330063459
  18. 18. Trojan J, Blossey BK, Johnson T, et al. Loss of tumorogenicity of rat glioblastoma directed by episome-based antisense cDNA transcription of insulin-like growth factor I. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(11):4874-4878
  19. 19. Le Roith D, Bondy C, Yakar S, Liu JL, Butler A. The somatomedin hypothesis. Endocrine Reviews. 2011;22(1):53-74. DOI: 10.1210/edrv.22.1.0419
  20. 20. Froesch CS, Schwander J, Zapf J. Actions of insulin-like growth factors. Annual Review of Physiology. 1985;47:443-467
  21. 21. Johnson TR, Trojan J, Rudin SD, et al. Effects on actinomycin D and cycloheximide on transcript levels of IGF- I, actin and albumin in hepatocyte primary cultures treated with growth hormone and insulin. Molecular Reproduction and Development. 1991;30:95-99
  22. 22. Trojan A, Jay LM, Kasprzak H, et al. Immunotherapy of malignant tumors using antisense anti-IGF-I approach: Case of glioblastoma. Journal of Cancer Therapy. 2014;5:685-705
  23. 23. Trojan J. Anti – Gene anti IGF-I technology applied for cancer immunotherapy. World Journal of Research Review. 2016;1(3):67-75
  24. 24. Trojan J, Uriel J. Immunocytochemical signaling of alpha fetoprotein (AFP) and serum-albumin (Alb) in ecto-, mesa- and endodermal tissue derivatives of the developing rat. Oncodevelopmental Biology and Medicine. 1982;3:12-22
  25. 25. Benno RH, Williams TH. Evidence for intracellular localization of AFP in the developing rat brain. Brain Research. 1978;142:182-186
  26. 26. Trojan J, Uriel J. Localisation intracellulaire of l’AFP et de la serum albumine dans le fœtal nerveux central du rat au cours du developpement fœtal et postnatal. Comptes Rendus de l’Académie des Sciences Paris. 1979;289:1157-1160
  27. 27. Trojan J, Johnson TR, Rudin SD, et al. Gene therapy of murine teratocarcinoma: Separate functions for insulin-like growth factors I and II in immunogenicity and differentiation. Proceedings of the National Academy of Sciences of the United States of America. 1994;91:6088-6092
  28. 28. Schubert D, Humphreys S, Baroni C, Cohn M. In vitro differentiation of a mouse neuroblastoma. Biochemistry. 1969;64:316-323
  29. 29. Bernal S, Thompson RA, Gilbert F, Baylin S. In vitro and in vivo growth characteristics of two different cell populations in an established line of human neuroblastoma. Cancer Research. 1983;43:125-164
  30. 30. Uriel J, Faivre-Baumann A, Trojan J, et al. Immunocytochemical demonstration of alphafoetoprotein uptake by primary cultures of foetal hemisphere cells from mouse brain. Neuroscience Letters. 1981;27:171-175
  31. 31. Uriel J, Poupon MF, Geuskens M. Alphaignaling uptake by cloned cell lines derived from a nickel-induced rat rhabdomyosarcoma. British Journal of Cancer. 1983;48:261-269
  32. 32. Uriel J, Villacampa MJ, Moro R, et al. Uptake of radiolabeled alphafetoprotein by mouse mammary carcinomas and usefulness in tumor scientigraphy. Cancer Research. 1984;44:5314-5319
  33. 33. Gaillard J, Caillaud JM, Maunoury R, et al. Expression du neuroectoblaste dans le teratocarcinome et le teratome de la souris. Bulletin de l'Institut Pasteur. 1984;82:335-385
  34. 34. Trojan J, Uriel J, Gaillard JA. Localisation de alphafoetoproteine dans les derives neuroepitheliaux des teratocarcinomes de la souris. Annales de Pathologie. 1983;3:137-145
  35. 35. Hajeri-Germond M, Trojan J, Uriel J, et al. In vitro uptake of exogenous alphafetoprotein by chicken dorsal root ganglia. Developmental Neuroscience. 1984;6:111-117
  36. 36. Castillo T, Trojan A, Noguera MC, et al. Epistemiologic experience in elaboration of molecular biology technology for immunogene therapy (in Spanish). Revista Cientifica. 2016;2:25. DOI: 10.14483/udistrital.jour.RC.2016.25.a6
  37. 37. Benedetti E, Galzio R, D’Angelo B, et al. PPARs in human neuroepithelial tumors: PPAR ligands as anticancer therapies for the most common human neuroepithelial tumors. PPAR Research. 2010:401-427. DOI: 10.1155/2010/427401
  38. 38. Lichtor T. Evolution of the Molecular Biology of Brain Tumors and the Therapeutic Implications. Vienna/Rijeka: InTech; 2013
  39. 39. Love S, Arie Perry A, Ironside J, Budka H. Greenfield’s Neuropathology. 9th ed. New York: CRC Press; 2015
  40. 40. Daughaday WH, Hall K, Raben MS, et al. Somatomedin: Proposed designation for sulphation factor. Nature. 1972;235:107
  41. 41. Han VKM, Hill DJ. In: Shofield PN, editor. The Insulin-Like Growth Factors: Structure and Biological Functions. Oxford, England: Oxford University Press; 1992. pp. 178-219
  42. 42. Baserga R, Sell C, Porcu P, Rubini M. The role of the IGF-I receptor in the growth and transformation of mammalian cells. Cell Proliferation. 1994;27:63-71
  43. 43. Baserga R. Oncogenes and the strategy of growth factors. Cell. 1994;79:927-930
  44. 44. Sturm MA, Conover CA, Pham H, et al. Insulin like growth factor receptors and binding proteins in rat neuroblastoma. Endocrinology. 1989;124:388-396
  45. 45. Werther GA, Abate M, Hogg A, et al. Localisation of Insulin like growth factor mRNA in rat brain by in situ hybridization-relation to IGF-I rceceptor. Molecular Endocrinology. 1990;4:773-778
  46. 46. Antoniades HN, Galanopoulis T, Nevile-Golden J, et al. Expression of insulin like growth factor I and II and their receptor mRNAs in primary human astrocytomas and meningiomas: In vivo studies using in situ hybridization and immunocytochemistry. International Journal of Cancer. 1992;50:215-222
  47. 47. Trojan J, Uriel J, Deugnier MA, et al. Immunocytochemical quantitative study of alpha-fetoprotein in normal and neoplastic neural development. Developmental Neuroscience. 1984;6(4-5):251-259. DOI: 10.1159/000112352
  48. 48. Ostos H, Astaiza G, Garcia F, et al. Decreased incidence of defects of neural tube closure at the University Hospital of Neiva: Possible effect promotion of folic acid (in Spanish). Biomédica. 2000;20(1):18-24. DOI: 10.7705/ignaling.v20i1.1043
  49. 49. Le Roith D. The insulin-like growth factor system. Experimental Diabesity Research. 2003;4(4):205-212. DOI: 10.1155/EDR.2003.205
  50. 50. Adhami VM, Afaq F, Mukhtar H. Insulin-like growth factor-I axis as a pathway for cancer chemoprevention. Clinical Cancer Research. 2006;12(19):5611-5614. DOI: 10.1158/1078-0432.CCR-06-1564
  51. 51. Chen H, Mester T, Raychaudhuri N, et al. Teprotumumab, an IGF-1R blocking monoclonal antibody inhibits TSH and IGF-1 action in fibrocytes. Journal of Clinical Endocrinology and Metabolism. 2014;99(9):E1635-E1640. DOI: 10.1210/jc.2014-1580
  52. 52. Kooijman R. Regulation of apoptosis by insulin-like growth factor (IGF)-I. Cytokine & Growth Factor Reviews. 2006;17(4):305-323. DOI: 10.1016/j.cytogfr.2006.02.002
  53. 53. Bueno S, Santandr R, Alvarez A, et al. Brain stem cells and IGF-I: Implications in development, regeneration and cancer therapeutics. Integrative Molecular Medicine. 2018;5(1). DOI: 10.15761/IMM.1000319
  54. 54. Kurmasheva RT, Houghton PJ. IGF-I mediated survival pathways in normal and malignant cells. Biochimica et Biophysica Acta. 2006;1766(1):1-22. DOI: 10.1016/j.bbcan.2006.05.003
  55. 55. Beckner ME, Gobbel GT, Abounader R, et al. Glycolytic glioma cells with active glycogen synthase are sensitive to PTEN and inhibitors of PI3K and gluconeogenesis. Laboratory Investigation. 2005;85(12):1457-1470. DOI: 10.1038/labinvest.3700355
  56. 56. Vignot S, Faivre S, Aguirre D, et al. mTOR-targeted therapy of cancer with rapamycin derivatives. Annals of Oncology. 2005;16(4):525-537
  57. 57. Schlingensiepen KH, Jaschinski F, Lang SA, et al. Transforming growth factor-beta 2 gene silencing with trabedersen (AP 12009) in pancreatic cancer. Cancer Science. 2011;102(6):1193-1200. DOI: 10.1111/j.1349-7006.2011.01917.x
  58. 58. Trojan J, Anthony DD. Antisense strategies in therapy of gliomas. Current Signal Transduction Therapy. 2011;6(3):411-423. DOI: 10.2174/157436211797483895
  59. 59. Patel S, Doble B, Woodgett JR. Glycogen synthase kinase-3 in insulin and Wnt signaling: A double-edged sword? Biochemical Society - Translation UK. 2004;32(5):803-808. DOI: 10.1042/BST0320803
  60. 60. Jiang R, Mircean C, Shmulevich I, et al. Pathway alterations during glioma progression revealed by reverse phase protein lysate arrays. Proteomics. 2006;6(10):2964-2971. DOI: 10.1002/pmic.200500555
  61. 61. Hutterer M, Gunsilius E, Stockhammer G. Molecular therapies for malignant glioma. Wien Medicinal Wochenschreibung. 2006;156(11):351-363. DOI: 10.1007/s10354-006-0308-3
  62. 62. Sachdev D, Yee D. Disrupting insulin-like growth factor signaling as a potential cancer therapy. Molecular Cancer Therapy. 2007;6(1):1-12. DOI: 10.1158/1535-7163.MCT-06-0080
  63. 63. Ardourel M, Blin M, Moret JL, et al. A new putative target for antisense gene therapy of glioma: Glycogen synthetase. Cancer Biology and Therapy. 2007;6(5):719-723. DOI: 10.4161/cbt.6.5.4232
  64. 64. Zhou X, Ren Y, Moore L, et al. Downregulation of miR-21 inhibits EGFR pathway and suppresses the growth of human glioblastoma cells independent of PTEN status. Laboratory Investigation. 2010;90:144-155. DOI: 10.1038/labinvest.2009.126
  65. 65. Premkumar DR, Arnold B, Jane EP, et al. Synergistic interaction between 17-AAG and phosphatidylinositol 3-kinase inhibition in human malignant glioma cells. Molecular Carcinogenesis. 2006;45(1):47-59. DOI: 10.1002/mc.20152
  66. 66. Sanson M, Laigle-Donadey F, Benouaich-Amiel A. Molecular changes in brain tumours: Prognostic and therapeutic impact. Current Opinion in Oncology. 2006;18(6):623-630. DOI: 10.1097/01.cco.0000245322.11787.72
  67. 67. Trojan J, Briceno I. IGF-I antisense and triple-helix gene therapy of glioblastoma. In: Pantar A, editor. Evolution of the Molecular Biology of Brain Tumors and the Therapeutic Implications. Vienna/Rijeka: InTech; 2013. pp. 149-166
  68. 68. Esiri M, Perl D. Oppenheimer’s Diagnostic Neuropathology. 3rd ed. FL: CRC Press; 2006
  69. 69. Glick RP, Lichtor T, Unterman TG. Insulin-like growth factors in central nervous system tumors. Journal of Neuro-Oncology. 1997;35(3):315-325
  70. 70. Obrepalska-Steplowska A, Kedzia A, Trojan J, et al. Analysis of coding and promoter sequences of the IGF-I gene in children with growth disorders presenting with normal level of growth hormone. Journal of Pediatric Endocrinology and Metabolism. 2003;16(9):1267-1275
  71. 71. Hu B, Niu X, Cheng L, et al. Discovering cancer biomarkers from clinical samples by protein microarrays. Proteomics: Clinical Applications. 2015;9(1-2):98-110. DOI: 10.1002/prca.201400094
  72. 72. Ertl DA, Gleiss A, Sagmeister S, et al. Determining the normal range for IGF-I, IGFBP-3, and ALS: New reference data based on current internal standards. Wiener Medizinische Wochenschrift. 2014;164(17-18):343-352. DOI: 10.1007/sl 0354-014-0299-4
  73. 73. Gu F, Schumacher FR, Canzian F, et al. Eighteen insulin-like growth factor pathway genes, circulating levels of IGF-I and its binding protein, and risk of prostate and breast cancer. Cancer Epidemiology, Biomarkers & Prevention. 2010;19(11):2877-2887. DOI: 10.1158/1055-9965.EPI-10-0507
  74. 74. Mosquera CJ, Castelblanco OL. Didactic questions and epistemiology (in Spanish). Revista Gondola. 2015;10(1). DOI: 10.14483/udistrital.jour.GDLA.2015.1.a00
  75. 75. Stupp R, Hegi ME, van den Bent MJ, et al. Changing paradigms—An update on the multidisciplinary management of malignant glioma. The Oncologist. 2006;11:165-180
  76. 76. Kjaergaard J, Wang L, Kuriyama H, Shu S, Plautz GE. Active immunotherapy for advanced intracranial murine tumors by using dendritic cell-tumor cell fusion vaccines. Journal of Neurosurgery. 2005;103:156-164
  77. 77. Rubenstein JL, Nicolas JF, Jacob F. Nonsense RNA: A tool for specifically inhibiting the expression of a gene in vivo. Comptes Rendus de l'Académie des Sciences, Paris III. 1984;299:271-274
  78. 78. Weintraub H, Izant J, Harland R. Antisense RNA as a molecular tool for genetic analysis. Trends in Genetics. 1985;1(1):23-25
  79. 79. Fakhrai H, Dorigo O, Shawler DL, et al. Eradication of established intracranial rat gliomas by transforming growth factor beta antisense gene therapy. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(7):2909-2914
  80. 80. Dervan P. Reagents for the sitespecific cleavage of megabase DNA. Nature. 1992;359:87-88
  81. 81. Hélène C. Control of oncogene expression by antisense nucleic acid. European Journal of Cancer. 1994;30A:1721-1726
  82. 82. Berezikov E, Thuemmler F, van Laake LW, et al. Diversity of microRNAs in human and chimpanzee brain. Nature Genetics. 2006;38(12):1375-1377
  83. 83. Corsten MF, Miranda R, Kasmieh R, et al. MicroRNA-21 knockdown disrupts glioma growth in vivo and displays synergistic cytotoxicity with neural precursor cell delivered S-TRAIL in human gliomas. Cancer Research. 2007;67(19):8994-9000
  84. 84. Boado RJ. RNA interference and nonviral targeted gene therapy of experimental brain cancer. NeuroRx. 2005;2(1):139-150
  85. 85. Pai SI, Lin YY, Macaes B, et al. Prospects of RNA interference therapy for cancer. Gene Therapy. 2006;13(6):464-477
  86. 86. Shevelev A, Burfeind P, Schulze E, et al. Potential triple helix mediated inhibition of IGF-I gene expression significantly reduces tumorigenicity of glioblastoma in an animal model. Cancer Gene Therapy. 1997;4:105-112
  87. 87. Rininsland F, Johnson T, Chernicky C, et al. 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 the National Academy of Sciences of the United States of America. 1997;94:5854-5859
  88. 88. Dias N, Stein CA. Basic concepts and antisense oligonucleotides mechanisms. Molecular Cancer Therapeutics. 2002;1:347-355
  89. 89. Wells DJ. Therapeutic restoration of dystrophin expression in Duchenne muscular dystrophy. Journal of Muscle Research and Cell Motility. 2006;27(5-7):387-398
  90. 90. Trojan J, Pan YX, Wei MX, et al. Methodology for anti-gene anti-IGF-I therapy of malignant tumours. Chemotherapy Research and Practice. 2011/2012. DOI: 10.1155/2012/721873
  91. 91. Townsend SE, Allison JA. Tumor rejection after direct costi-mulation of CD8+ T cell transfected melanoma cells. Science 1993;259: 368-370
  92. 92. Guo Y, Wu M, Chen H, et al. Effective tumor vaccine generated by fusion of hepatoma cells with lymphocytes B cells. Science. 1994;263:518-520
  93. 93. Trojan J. Cancer Immunogene Therapy. Anti-Gene Anti IGF-I Approach. Case of Glioblastoma. Germany: Lambert Academic Publishers; 2017. 140p. ISBN-13: 978-3-330-06345-7; ISBN-10: 3330063459
  94. 94. Linsley PS, Clark EA, Ledbetter JA. T-cell antigen CD28 mediates adhesion with B cells by interacting with ativation antigen B7/13. Proceedings of the National Academy of Sciences of the United States of America. 1990;87:5031-5035
  95. 95. Freeman GB, Gray GS, Gimmi CD, et al. Structure
  96. 96. Upegui-Gonzalez LC, Ly A, Sierzega M, et al. IGF-I triple helix strategy in hepatoma treatment. Hepato-Gastroenterology. 2001;48:660-666
  97. 97. Steiman RM, Turkey S, Mellman I, Inaba K. The induction of tolerance by dendritic cells that have captured apoptotic cells. Journal of Experimental Medicine. 2000;191:411-416
  98. 98. Lafarge-Frayssinet C, Sarasin A, Duc HT, et al. Gene therapy for hepatocarcinoma: Antisense IGF-I transfer into a rat hepatoma cell line inhibits tumorigenesis into syngeneic animal. Cancer Gene Therapy. 1997;4:276-285
  99. 99. Schwartz RH. Costimulation of T lymphocytes: The role of CD28, CTLA-4 and B7/BBI in interleukin-2 production and immunotherapy. Cell. 1992;71:1065-1068
  100. 100. D’Ambrosio C, Ferber A, Resnicoff M, et al. A soluble insulin-like growth factor receptor that induces apoptosis of tumor cells in vivo and inhibits tumorigenesis. Cancer Research. 1996;56:4013-4020
  101. 101. Dudek H, Datta SR, Franke TS, et al. Regulation of neuronal servival by the serine-threonine protein kinase. Akta Science. 1997;275:661-665
  102. 102. Satoh J, Lee YB, Kim SU. T-cell costimulatory molecules B7-1 (CD80) and B7-2 (CD86) are expressed in human microglia but not in astrocytes in culture. Brain Research. 1995;704:95-96
  103. 103. Resnicoff M, Abraham D, Yutanawiboonchai W, et al. The insulin-like growth factor I receptor protects tumor cells from apoptosis in vitro. Cancer Research. 1995;55:2463-2469
  104. 104. Sotomayor E, Fu Y, Lopez-Cepero M, et al. Role of tumor derived cytokines on the immune system of mice bearing a mammary adenocarcinoma. Journal of Immunology. 1991;147:2816-2823
  105. 105. Matthew L, Saiter B, Bhardwag N. Dendritic cells acquire antigen from apoptotic cells and induce class I restricted CTL. Nature. 1998;392:86-89
  106. 106. Chen L, Ashe S, Brady WA, et al. Costimulation of anti-tumor immunity by the B7 counter receptor for the T lymphocyte molecules CD28 and CTLA-4. Cell. 1992;71:1093-1102
  107. 107. Anthony D, Pan Y, Wu S, et al. Ex vivo and in vivo IGF-I antisense RNA strategies for treatment of cancer in humans. Advances in Experimental Medicine and Biology. 1998;451:27-34
  108. 108. Zhu C, Trabado S, Fan Y, et al. Characterization of effector components from the humoral and cellular immune response stimulated by melanoma cells exhibiting modified IGF-1 expression. Biomedicine & Pharmacotherapy. 2015;70:53-57. DOI: 10.1016/j.biopha.2015.01.002
  109. 109. Gerard CM, Bruyns C, Delvaux A, et al. Loss of tumorigenicity and increased immunogenicity induced by interleukin-10 gene transfer in B16 melanoma cells. Human Gene Therapy. 1996;7(1):23-31
  110. 110. Buck C, Digel W, Schoniger W, et al. Tumor nercrosis factor alpha but not lymphotoxin, stimulates growth of tumor cells in hairy cell leukemia. Leukemia. 1990;4:431-439
  111. 111. Aggarwal B, Schwarz L, Hogan M, et al. Triple helixforming oligodeoxiribonucleotides 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
  112. 112. Blanchet O, Bourge JF, Zinszner H, et al. Altered binding of regulatory factors to HLA class I enhancer sequence in human tumor cell lines lacking class I antigen expression. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(8):3488-3492
  113. 113. Trojan LA, Ly A, Kopinski P, et al. Antisense and triple helix anti IGF-I tumour vaccines: Gene therapy of gliomas. International Journal of Cancer Prevention. 2007;2(4):227-243
  114. 114. Wongkajornslip A, Ouyprasertkul M, Sangruchi T, et al. The analysis of peri-tumor necrosis following the subcutaneous implantation of autologous tumor cells transfected with an episome transcribing an antisense insulin-like growth factor I RNA in a glioblastoma multiforme subject. Journal of the Medical Association of Thailand. 2001;m4(3):740-747
  115. 115. Trojan LA, Kopinski P, Mazurek A, et al. IGF I triple helix gene therapy of rat and human gliomas. Annales Academiae Medicae Bialostocensis. 2003;48:18-27
  116. 116. Culver KW, Rarn Z, Wallbridge S. In vivo gene transfer with retroviral vector-producer celIs for treatment of experimental brain tumors. Science. 1992;256:1550-1552
  117. 117. Kopinski P, Ly A, Trojan J. Chapter 44: Antisense in oncology. In: Khayat D, Ly A, editors. About Cancer in Africa. Paris, France: Springer; 2005/2006
  118. 118. Grossman SA, Alavi JB, Supko JG, et al. Efficacy and toxicity of the antisense oligonucleotide aprinocarsen directed against protein kinase C-alpha delivered as a 21-day continuous intravenous infusion in patients with recurrent high-grade astrocytomas. Neuro-Oncology. 2005;7(1):32-40
  119. 119. Bernard-Helary K, Ardourel M, Magistrett P, et al. Stable transfection of cDNAs targeting specific steps of glycogen metabolism supports the existence of active gluconeogenesis in mouse cultured astrocytes. Glia. 2002;37(4):379-382
  120. 120. Andrews DW, Resnicoff M, Flanders A, et al. 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
  121. 121. Samani AA, Fallavollita L, Jaalouk DE, et al. Inhibition of carcinoma cell growth and metastasis by a vesicular stomatitis virus G-pseudotyped retrovector expressing type 1 insulin-like growth factor receptor antisense. Human Gene Therapy. 2001;12:1969-1977
  122. 122. Schlingensiepen R, Goldbrunner M, Szyrach MNI, et al. Intracerebral and intrathecal infusion of the TGF-beta2-specific antisense phosphorothioate oligonucleotide AP 12009 in rabbits and primates: Toxicology and safety. Oligonucleotides. 2005;15(2):94-104
  123. 123. Schlingensiepen KH, Schlingensiepen R, Steinbrecher A, et al. Targeted tumor therapy with the TGF-beta2 antisense compound AP 12009. Cytokine & Growth Factor Reviews. 2006;17:129-139
  124. 124. Hau P, Jachimczak P, Schlingensiepen R, et al. Inhibition of TGF-beta2 with AP 12009 in recurrent malignant gliomas: From preclinical to phase I/II studies. Oligonucleotides. 2007;17(2):201-212
  125. 125. Schlingensiepen KH, Fischer-Blass B, Schmaus S, et al. Antisense therapeutics for tumor treatment: the TGF-beta2 inhibitor AP 12009 in clinical development against malignant tumors. Recent Results in Cancer Research. 2008;177:137-150
  126. 126. Anthony DD. Ex vivo and in vivo IGF-1 antisense RNA strategies for treatment of cancers in humans [Abstract]. Cancer Gene Therapy. 1997;2(6):S322
  127. 127. Popiela T, Sierzega M, Gach T, et al. Phase I trial of colorectal cancer immunotherapy using autologous cancer cells transfected with an IGF-I antisense plasmid [abstract]. Acta Chirurgica Belgica. 2003;5(103):S2-S3
  128. 128. Schlingensiepen R, Goldbrunner M, Bischof A, et al. Antisense-TGF-beta-2 oligonucleotide AP 12009: Results of safety pharmacology and toxicity studies [abstract]. Journal of Cancer Research and Clinical Oncology. 2002;128(1):S134
  129. 129. Sierzega M, Jarocki P, Trojan J, et al. Gene immunotherapy of pancreatic cancer using IGF-I antisense approach; preliminary results [Abstract]. Journal of Hepato-Biliary-Pancreatic Surgery. 2002;9(1):S226
  130. 130. Trojan J, Kopinski P, Drewa T, et al. Immunogenotherapy of prostate cancer. Urologia Polska. 2003;56(2):7-11
  131. 131. Messaoudi S. Recent advances in Hsp90 inhibitors as antitumour agents. Anti-Cancer Agents in Medicinal Chemistry. 2008;8:761-782
  132. 132. Goudar RK, Shi Q, Hjelmeland MD, et al. 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
  133. 133. Lamszus K, Brockman MA, Eckerich C, et al. Inhibition of glioblastoma angiogenesis and invasion by combined treatments directed against vascular endothelial growth factor receptor-2, epidermal growth factor receptor, and vascular endothelial-cadherin. Clinical Cancer Research. 2005;11:4934-4940
  134. 134. Reardon DA, Quinn JA, Vredenburgh JJ, et al. Phase 1 trial of gefitinib plus sirolimus in adults with recurrent malignant glioma. Clinical Cancer Research. 2006;12:860-868
  135. 135. Halatsch ME, Schmidt U, Behnke-Mursch L, Untenberg A, Wirtz CR. Epidermal growth factor inhibition for the treatment of glioblastoma multiforme and other malignant brain tumours. Cancer Treatment Reviews. 2006;32:74-89
  136. 136. Wen PY, Yung WK, Lamborn KR, et al. Phase I/II study of imatinib mesylate for recurrent malignant gliomas: Norty American Brain Tumour Consortium Study 99-08. Clinical Cancer Research. 2006;12:4899-4907
  137. 137. 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
  138. 138. Trojan LA, Ly A, Upegui-Gonzalez LC, et al. Antisense anti IGF-I therapy of primary hepatic cancer. Journal Africain du Cancer. 2009;1:1-10
  139. 139. Biroccio A, Leonett C, Zupi G. The future of antisense therapy: Combination with anticancer treatment. Oncogene. 2003;22:6579-6588
  140. 140. Meng Y, Carpentier AF, Chen L, et al. Successful combination of local CpG-ODN and radiotherapy in malignant glioma. International Journal of Cancer. 2005;116:992-997
  141. 141. Gonzalez J, Gilbert MR. Treatment of astrocytomas. Current Opinion in Neurology. 2005;18:632-638
  142. 142. Jane EP, Premkumar DR, Pollack IF. Coadministration of sorafenib with rottlerin potently inhibits cell proliferation and migration in human malignant glioma cells. Journal of Pharmacology and Experimental Therapy. 2006;319:1070-1080
  143. 143. Vega EA, Graner MW, Sampson JH. Combating immunosuppression in glioma. Future Oncology. 2008;4(3):433-442
  144. 144. Trojan J. Brain - From Development to Neoplasia and Gene Therapy Solution. Germany: Lambert Academic Publishers; 2018. 150p. ISBN: 978-620-2-08024-8
  145. 145. Dietrich PY, Dutoit V, Tran Thang NN, et al. T cell immunotherapy for malignant glioma: Toward a combined approach. Current Opinion in Oncology. 2010;22(6):604-610
  146. 146. Wikipedia—Gene Therapy, History 1990s–2010s
  147. 147. Trojan A, Aristizabal B, Jay LM, et al. Testing of IGF-I biomarker in an ethical context. Advances in Modern Oncology Research. 2016;2(4). DOI: 10.18282/amor:v2:i4.58

Written By

Trojan Jerzy

Submitted: 31 July 2018 Reviewed: 15 November 2018 Published: 15 March 2019