open access

Vol 74, No 6 (2023)
Review paper
Submitted: 2023-05-17
Accepted: 2023-10-03
Published online: 2023-12-05
Get Citation

A new antiviral hypothesis and radioactive iodine therapy to other cancers, such as breast cancer, lung cancer, and glioblastoma multiforme (GBM)?

Agata Czarnywojtek12, Paweł Gut2, Magdalena Borowska1, Nadia Sawicka-Gutaj2, Paweł Caputa3, Beata Kos-Kudła4, Marek Ruchała2, Marzena Dworacka5
·
Endokrynol Pol 2023;74(6).
Affiliations
  1. Department of Pharmacology, Poznan University of Medical Sciences, Poznan, Poland
  2. Chair and Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
  3. Department of Oncology, Greater Poland Cancer Centre, Poznan, Poland
  4. Department of Endocrinology and Neuroendocrine Tumours, Department of Pathophysiology and Endocrinology, Medical University of Silesia, Katowice, Poland
  5. Coordinator of the International Head and Neck Scientific Group, Padua, Italy

open access

Vol 74, No 6 (2023)
Review Article
Submitted: 2023-05-17
Accepted: 2023-10-03
Published online: 2023-12-05

Abstract

Radioactive iodine therapy (RIT) is an effective, safe, and cheap method in benign and malignant thyroid diseases. There is still an unresolved question of whether RIT treatment also plays a role in the treatment of, for example, breast cancer, lung cancer, or glioblastoma multiforme (GBM). These studies are currently being carried out in rats in combination with genes, but it may be an interesting challenge to assess “pure” RIT alone, thanks to the expression of sodium iodide symporter (NIS), is effective in other organ nodules, both benign and malignant. Cloning of the NIS in 1996 provided an opportunity to use NIS as a powerful theranostic transgene. In addition, NIS is a sensitive reporter gene that can be monitored by high-resolution PET imaging using the radiolabels [124I]sodium iodide ([124I]NaI) or [18F] tetrafluoroborate ([18F]TFB). Based on published positron emission tomography (PET) results, [124I]sodium iodide and internally synthesized [18F]TFB were compared in an orthotopic animal model of NIS-expressing glioblastoma. The results showed improved image quality using [18F]TFB. Based on these results, we will be able to extend the NIS gene therapy approach using non-viral gene delivery vehicles to target orthotopic tumour models with low-volume disease such as GBM. Is it possible to treat RIT alone without using the NIS gene in GBM? After all, the NIS symporter was detected not only in the thyroid gland, but also in different tumours. The administration of RIT is completely harmless; the only complication is hypothyroidism. Indeed, recently it has been shown that, for example, in the case of thyroid cancer, the maximum RIT is 37000 MBq (1000 mCi). When beneficial effects of therapy in GBM are not possible (e.g. neurosurgery, modulated electro-hyperthermia, chemotherapy, immunotherapy, cancer vaccines, or oncolytic viruses), could RIT provide a “revolution” using NIS?

Abstract

Radioactive iodine therapy (RIT) is an effective, safe, and cheap method in benign and malignant thyroid diseases. There is still an unresolved question of whether RIT treatment also plays a role in the treatment of, for example, breast cancer, lung cancer, or glioblastoma multiforme (GBM). These studies are currently being carried out in rats in combination with genes, but it may be an interesting challenge to assess “pure” RIT alone, thanks to the expression of sodium iodide symporter (NIS), is effective in other organ nodules, both benign and malignant. Cloning of the NIS in 1996 provided an opportunity to use NIS as a powerful theranostic transgene. In addition, NIS is a sensitive reporter gene that can be monitored by high-resolution PET imaging using the radiolabels [124I]sodium iodide ([124I]NaI) or [18F] tetrafluoroborate ([18F]TFB). Based on published positron emission tomography (PET) results, [124I]sodium iodide and internally synthesized [18F]TFB were compared in an orthotopic animal model of NIS-expressing glioblastoma. The results showed improved image quality using [18F]TFB. Based on these results, we will be able to extend the NIS gene therapy approach using non-viral gene delivery vehicles to target orthotopic tumour models with low-volume disease such as GBM. Is it possible to treat RIT alone without using the NIS gene in GBM? After all, the NIS symporter was detected not only in the thyroid gland, but also in different tumours. The administration of RIT is completely harmless; the only complication is hypothyroidism. Indeed, recently it has been shown that, for example, in the case of thyroid cancer, the maximum RIT is 37000 MBq (1000 mCi). When beneficial effects of therapy in GBM are not possible (e.g. neurosurgery, modulated electro-hyperthermia, chemotherapy, immunotherapy, cancer vaccines, or oncolytic viruses), could RIT provide a “revolution” using NIS?

Get Citation

Keywords

sodium iodide symporter (NIS); glioblastoma multiforme (GM); gene therapy, RIT radioiodine treatment (RIT)

About this article
Title

A new antiviral hypothesis and radioactive iodine therapy to other cancers, such as breast cancer, lung cancer, and glioblastoma multiforme (GBM)?

Journal

Endokrynologia Polska

Issue

Vol 74, No 6 (2023)

Article type

Review paper

Published online

2023-12-05

Page views

534

Article views/downloads

260

DOI

10.5603/ep.95505

Bibliographic record

Endokrynol Pol 2023;74(6).

Keywords

sodium iodide symporter (NIS)
glioblastoma multiforme (GM)
gene therapy
RIT radioiodine treatment (RIT)

Authors

Agata Czarnywojtek
Paweł Gut
Magdalena Borowska
Nadia Sawicka-Gutaj
Paweł Caputa
Beata Kos-Kudła
Marek Ruchała
Marzena Dworacka

References (114)
  1. Luster M, Pfestroff A, Hänscheid H, et al. Radioiodine Therapy. Semin Nucl Med. 2017; 47(2): 126–134.
  2. Levy O, Dai G, Riedel C, et al. Cloning and characterization of the thyroid iodide transporter. Nature. 1996; 379(6564): 458–460.
  3. Spitzweg C, Joba W, Eisenmenger W, et al. Analysis of human sodium iodide symporter gene expression in extrathyroidal tissues and cloning of its complementary deoxyribonucleic acids from salivary gland, mammary gland, and gastric mucosa. J Clin Endocrinol Metab. 1998; 83(5): 1746–1751.
  4. Carrasco N. Thyroid iodide transport: the Na+ /J – symporter (NIS). In: Braveman LE, Utiger RD. ed. Werner & Ingbar’s the thyroid: a fundamental and clinical text. Lippincott Wiliams & Wilkins, Philadelphia 2000: Philadelphia.
  5. De La Vieja A, Dohan O, Levy O, et al. Molecular analysis of the sodium/iodide symporter: impact on thyroid and extrathyroid pathophysiology. Physiol Rev. 2000; 80(3): 1083–1105.
  6. Hingorani M, Spitzweg C, Vassaux G, et al. The biology of the sodium iodide symporter and its potential for targeted gene delivery. Curr Cancer Drug Targets. 2010; 10(2): 242–267.
  7. Llorente-Esteban A, Manville RW, Reyna-Neyra A, et al. Allosteric regulation of mammalian Na/I symporter activity by perchlorate. Nat Struct Mol Biol. 2020; 27(6): 533–539.
  8. Spitzweg C, Bible KC, Hofbauer LC, et al. Advanced radioiodine-refractory differentiated thyroid cancer: the sodium iodide symporter and other emerging therapeutic targets. Lancet Diabetes Endocrinol. 2014; 2(10): 830–842.
  9. Spitzweg C, Nelson PJ, Wagner E, et al. The sodium iodide symporter (NIS): novel applications for radionuclide imaging and treatment. Endocr Relat Cancer. 2021; 28(10): T193–T213.
  10. Willhauck MJ, Sharif Samani BR, Gildehaus FJ, et al. Application of 188rhenium as an alternative radionuclide for treatment of prostate cancer after tumor-specific sodium iodide symporter gene expression. J Clin Endocrinol Metab. 2007; 92(11): 4451–4458.
  11. Willhauck MJ, Samani BRS, Wolf I, et al. The potential of 211Astatine for NIS-mediated radionuclide therapy in prostate cancer. Eur J Nucl Med Mol Imaging. 2008; 35(7): 1272–1281.
  12. Dadachova E, Bouzahzah B, Zuckier LS, et al. Rhenium-188 as an alternative to Iodine-131 for treatment of breast tumors expressing the sodium/iodide symporter (NIS). Nucl Med Biol. 2002; 29(1): 13–18.
  13. Shimura H, Haraguchi K, Miyazaki A, et al. Iodide uptake and experimental 131I therapy in transplanted undifferentiated thyroid cancer cells expressing the Na+/I- symporter gene. Endocrinology. 1997; 138(10): 4493–4496.
  14. Levy O, Vieja AD, Ginter C, et al. N-linked Glycosylation of the Thyroid Na+/I− Symporter (NIS). J Biol Chem. 1998; 273(35): 22657–22663.
  15. Mian C, Lacroix L, Alzieu L, et al. Sodium iodide symporter and pendrin expression in human thyroid tissues. Thyroid. 2001; 11(9): 825–830.
  16. Jhiang SM, Cho JY, Ryu KY, et al. An immunohistochemical study of Na+/I- symporter in human thyroid tissues and salivary gland tissues. Endocrinology. 1998; 139(10): 4416–4419.
  17. Wapnir IL, van de Rijn M, Nowels K, et al. Immunohistochemical profile of the sodium/iodide symporter in thyroid, breast, and other carcinomas using high density tissue microarrays and conventional sections. J Clin Endocrinol Metab. 2003; 88(4): 1880–1888.
  18. Joba W, Spitzweg C, Schriever K, et al. Analysis of human sodium/iodide symporter, thyroid transcription factor-1, and paired-box-protein-8 gene expression in benign thyroid diseases. Thyroid. 1999; 9(5): 455–466.
  19. Kogai T, Endo T, Saito T, et al. Regulation by thyroid-stimulating hormone of sodium/iodide symporter gene expression and protein levels in FRTL-5 cells. Endocrinology. 1997; 138(6): 2227–2232.
  20. Venkataraman GM, Yatin M, Ain KB. Cloning of the human sodium-iodide symporter promoter and characterization in a differentiated human thyroid cell line, KAT-50. Thyroid. 1998; 8(1): 63–69.
  21. Smyth P, Dwyer RM. The sodium iodide symporter and thyroid disease. Commentary. Clin Endocrinol 2002; 56:427–429. .109/thy.199. Clin Endocrinol. 2002; 56(4): 427–429.
  22. Riedel C, Levy O, Carrasco N. Post-transcriptional regulation of the sodium/iodide symporter by thyrotropin. J Biol Chem. 2001; 276(24): 21458–21463.
  23. Chung JK. Sodium iodide symporter: its role in nuclear medicine. J Nucl Med. 2002; 43(9): 1188–1200.
  24. Uyttersprot N, Pelgrims N, Carrasco N, et al. Moderate doses of iodide in vivo inhibit cell proliferation and the expression of thyroperoxidase and Na+/I- symporter mRNAs in dog thyroid. Mol Cell Endocrinol. 1997; 131(2): 195–203.
  25. Eng PH, Cardona GR, Previti MC, et al. Regulation of the sodium iodide symporter by iodide in FRTL-5 cells. Eur J Endocrinol. 2001; 144(2): 139–144.
  26. Scipioni A, Ferretti E, Soda G, et al. hNIS protein in thyroid: the iodine supply influences its expression and localization. Thyroid. 2007; 17(7): 613–618.
  27. Kohn LD, Suzuki K, Nakazato M, et al. Effects of thyroglobulin and pendrin on iodide flux through the thyrocyte. Trends Endocrinol Metab. 2001; 12(1): 10–16.
  28. Suzuki K, Mori A, Saito J, et al. Follicular thyroglobulin suppresses iodide uptake by suppressing expression of the sodium/iodide symporter gene. Endocrinology. 1999; 140(11): 5422–5430.
  29. Wolff J, Chaikoff IL. Plasma inorganic iodide as a homeostatic regulator of thyroid function. J Biol Chem. 1948; 174(2): 555–564.
  30. Wolff J, Chaikoff IL. The temporary nature of the inhibitory action of excess iodine on organic iodine synthesis in the normal thyroid. Endocrinology. 1949; 45(5): 504–13, illust.
  31. Spitzweg C, Joba W, Morris JC, et al. Regulation of sodium iodide symporter gene expression in FRTL-5 rat thyroid cells. Thyroid. 1999; 9(8): 821–830.
  32. Eng PH, Cardona GR, Fang SL, et al. Escape from the acute Wolff-Chaikoff effect is associated with a decrease in thyroid sodium/iodide symporter messenger ribonucleic acid and protein. Endocrinology. 1999; 140(8): 3404–3410.
  33. Spitzweg C, Morris JC. The sodium iodide symporter: its pathophysiological and therapeutic implications. Clin Endocrinol (Oxf). 2002; 57(5): 559–574.
  34. Ajjan RA, Kamaruddin NA, Crisp M, et al. Regulation and tissue distribution of the human sodium iodide symporter gene. Clin Endocrinol (Oxf). 1998; 49(4): 517–523.
  35. Ajjan RA, Watson PF, Findlay C, et al. The sodium iodide symporter gene and its regulation by cytokines found in autoimmunity. J Endocrinol. 1998; 158(3): 351–358.
  36. Morshed SA, Davies TF. Graves' Disease Mechanisms: The Role of Stimulating, Blocking, and Cleavage Region TSH Receptor Antibodies. Horm Metab Res. 2015; 47(10): 727–734.
  37. Ajjan RA, Findlay C, Metcalfe RA, et al. The modulation of the human sodium iodide symporter activity by Graves' disease sera. J Clin Endocrinol Metab. 1998; 83(4): 1217–1221.
  38. Tuttle R, Patience T, Budd S. Treatment with Propylthiouracil before Radioactive Iodine Therapy Is Associated with a Higher Treatment Failure Rate Than Therapy with Radioactive Iodine Alone in Graves' Disease. Thyroid. 1995; 5(4): 243–247.
  39. Sabri O, Zimny M, Schreckenberger M, et al. Success rate of radioiodine therapy in Graves' disease: the influence of thyrostatic medication. J Clin Endocrinol Metab. 1999; 84(4): 1229–1233.
  40. Sabri O, Zimny M, Schulz G, et al. Success rate of radioiodine therapy in Graves' disease: the influence of thyrostatic medication. J Clin Endocrinol Metab. 1999; 84(4): 1229–1233.
  41. Körber C, Schneider P, Körber-Hafner N, et al. Antithyroid drugs as a factor influencing the outcome of radioiodine therapy in Graves' disease and toxic nodular goitre? Eur J Nucl Med. 2001; 28(9): 1360–1364.
  42. Furlanetto TW, Nunes RB, Sopelsa AM, et al. Estradiol decreases iodide uptake by rat thyroid follicular FRTL-5 cells. Braz J Med Biol Res. 2001; 34(2): 259–263.
  43. Pekary AE, Hershman JM. Tumor necrosis factor, ceramide, transforming growth factor-beta1, and aging reduce Na+/I- symporter messenger ribonucleic acid levels in FRTL-5 cells. Endocrinology. 1998; 139(2): 703–712.
  44. Louis DN, Perry A, Wesseling P, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. 2016; 131(6): 803–820.
  45. Guo R, Xi Y, Zhang M, et al. Human sodium iodide transporter gene-mediated imaging and therapy of mouse glioma, comparison between Re and I. Oncol Lett. 2018; 15(3): 3911–3917.
  46. Cho JY, Shen DHY, Yang W, et al. In vivo imaging and radioiodine therapy following sodium iodide symporter gene transfer in animal model of intracerebral gliomas. Gene Ther. 2002; 9(17): 1139–1145.
  47. Opyrchal M, Allen C, Msaouel P, et al. Effective radiovirotherapy for malignant gliomas by using oncolytic measles virus strains encoding the sodium iodide symporter (MV-NIS). Hum Gene Ther. 2012; 23(4): 419–427.
  48. Ravera S, Reyna-Neyra A, Ferrandino G, et al. The Sodium/Iodide Symporter (NIS): Molecular Physiology and Preclinical and Clinical Applications. Annu Rev Physiol. 2017; 79: 261–289.
  49. Jiang H, Bansal A, Pandey MK, et al. Synthesis of 18F-Tetrafluoroborate via Radiofluorination of Boron Trifluoride and Evaluation in a Murine C6-Glioma Tumor Model. J Nucl Med. 2016; 57(9): 1454–1459.
  50. Kitzberger C, Spellerberg R, Morath V, et al. The sodium iodide symporter (NIS) as theranostic gene: its emerging role in new imaging modalities and non-viral gene therapy. EJNMMI Res. 2022; 12(1): 25.
  51. Niess H, von Einem JC, Thomas MN, et al. Treatment of advanced gastrointestinal tumors with genetically modified autologous mesenchymal stromal cells (TREAT-ME1): study protocol of a phase I/II clinical trial. BMC Cancer. 2015; 15: 237.
  52. Khoshnevisan A, Jauregui-Osoro M, Shaw K, et al. [(18)F]tetrafluoroborate as a PET tracer for the sodium/iodide symporter: the importance of specific activity. EJNMMI Res. 2016; 6(1): 34.
  53. Portulano C, Paroder-Belenitsky M, Carrasco N. The Na+/I- symporter (NIS): mechanism and medical impact. Endocr Rev. 2014; 35(1): 106–149.
  54. Spellerberg R, Benli-Hoppe T, Kitzberger C, et al. Selective sodium iodide symporter () genetherapy of glioblastoma mediatedby EGFR-targeted lipopolyplexes. Mol Ther Oncolytics. 2021; 23: 432–446.
  55. Vadysirisack DD, Shen DH, Jhiang SM. Correlation of Na+/I- symporter expression and activity: implications of Na+/I- symporter as an imaging reporter gene. J Nucl Med. 2006; 47(1): 182–190.
  56. Huang M, Batra RK, Kogai T, et al. Ectopic expression of the thyroperoxidase gene augments radioiodide uptake and retention mediated by the sodium iodide symporter in non-small cell lung cancer. Cancer Gene Ther. 2001; 8(8): 612–618.
  57. Dingli D, Bergert ER, Bajzer Z, et al. Dynamic iodide trapping by tumor cells expressing the thyroidal sodium iodide symporter. Biochem Biophys Res Commun. 2004; 325(1): 157–166.
  58. Spitzweg C, Dietz AB, O'Connor MK, et al. In vivo sodium iodide symporter gene therapy of prostate cancer. Gene Ther. 2001; 8(20): 1524–1531.
  59. Elisei R, Vivaldi A, Ciampi R, et al. Treatment with drugs able to reduce iodine efflux significantly increases the intracellular retention time in thyroid cancer cells stably transfected with sodium iodide symporter complementary deoxyribonucleic acid. J Clin Endocrinol Metab. 2006; 91(6): 2389–2395.
  60. AMYES EW, DEEB PH, VOGEL PJ, et al. Determining the site of brain tumors; the use of radioactive iodine and phosphorus. Calif Med. 1955; 82(3): 167–170.
  61. Yang J, Zheng R, Liang M, et al. Association of the Cumulative Dose of Radioactive Iodine Therapy With Overall Survival in Patients With Differentiated Thyroid Cancer and Pulmonary Metastases. Front Oncol. 2019; 9: 558.
  62. Dorn R, Kopp J, Vogt H, et al. Dosimetry-guided radioactive iodine treatment in patients with metastatic differentiated thyroid cancer: largest safe dose using a risk-adapted approach. J Nucl Med. 2003; 44(3): 451–456.
  63. Ross DS, Burch HB, Cooper DS, et al. 2016 American Thyroid Association Guidelines for Diagnosis and Management of Hyperthyroidism and Other Causes of Thyrotoxicosis. Thyroid. 2016; 26(10): 1343–1421.
  64. Wartofsky L, Glinoer D, Solomon B, et al. Differences and similarities in the diagnosis and treatment of Graves' disease in Europe, Japan, and the United States. Thyroid. 1991; 1(2): 129–135.
  65. Kirkpatrick DB. The first primary brain-tumor operation. J Neurosurg. 1984; 61(5): 809–813.
  66. Parvez K, Parvez A, Zadeh G. The diagnosis and treatment of pseudoprogression, radiation necrosis and brain tumor recurrence. Int J Mol Sci. 2014; 15(7): 11832–11846.
  67. Ellingson BM, Chung C, Pope WB, et al. Pseudoprogression, radionecrosis, inflammation or true tumor progression? challenges associated with glioblastoma response assessment in an evolving therapeutic landscape. J Neurooncol. 2017; 134(3): 495–504.
  68. Fiorentini G, Szasz A. Hyperthermia today: electric energy, a new opportunity in cancer treatment. J Cancer Res Ther. 2006; 2(2): 41–46.
  69. Alifieris C, Trafalis DT. Glioblastoma multiforme: Pathogenesis and treatment. Pharmacol Ther. 2015; 152: 63–82.
  70. Baxi S, Yang A, Gennarelli RL, et al. Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis. BMJ. 2018; 360: k793.
  71. Reardon DA, Kim TM, Frenel JS, et al. Treatment with pembrolizumab in programmed death ligand 1-positive recurrent glioblastoma: Results from the multicohort phase 1 KEYNOTE-028 trial. Cancer. 2021; 127(10): 1620–1629.
  72. Aurisicchio L, Pallocca M, Ciliberto G, et al. The perfect personalized cancer therapy: cancer vaccines against neoantigens. J Exp Clin Cancer Res. 2018; 37(1): 86.
  73. Palande V, Siegal T, Detroja R, et al. Detection of gene mutations and gene-gene fusions in circulating cell-free DNA of glioblastoma patients: an avenue for clinically relevant diagnostic analysis. Mol Oncol. 2022; 16(10): 2098–2114.
  74. Gleevec (imatinib mesylate) tablet. Daily Med; U.S. National Library of Medicine.
  75. Sun Li, Liang C, Shirazian S, et al. Discovery of 5-[5-fluoro-2-oxo-1,2- dihydroindol-(3Z)-ylidenemethyl]-2,4- dimethyl-1H-pyrrole-3-carboxylic acid (2-diethylaminoethyl)amide, a novel tyrosine kinase inhibitor targeting vascular endothelial and platelet-derived growth factor receptor tyrosine kinase. J Med Chem. 2003; 46(7): 1116–1119.
  76. Strumberg D. Preclinical and clinical development of the oral multikinase inhibitor sorafenib in cancer treatment. Drugs Today (Barc). 2005; 41(12): 773–784.
  77. Murphy DA, Makonnen S, Lassoued W, et al. Inhibition of tumor endothelial ERK activation, angiogenesis, and tumor growth by sorafenib (BAY43-9006). Am J Pathol. 2006; 169(5): 1875–1885.
  78. König D, Hench J, Frank S, et al. Larotrectinib Response in NTRK3 Fusion-Driven Diffuse High-Grade Glioma. Pharmacology. 2022; 107(7-8): 433–438.
  79. Subbiah V, Velcheti V, Tuch BB, et al. Selective RET kinase inhibition for patients with RET-altered cancers. Ann Oncol. 2018; 29(8): 1869–1876.
  80. https://www.magforce.com/news/?article=375.
  81. Schirrmacher V. Cancer Vaccines and Oncolytic Viruses Exert Profoundly Lower Side Effects in Cancer Patients than Other Systemic Therapies: A Comparative Analysis. Biomedicines. 2020; 8(3).
  82. Schirrmacher V, Lorenzen D, Van Go, et al. A new strategy of cancer immunotherapy combining hyperthermia/oncolytic virus pretreatment with specific autologous anti-tumor vaccination — A Review. Austin Oncol Case Rep. 2017; 2: 1006.
  83. Lowenfeld L, Mick R, Datta J, et al. Dendritic Cell Vaccination Enhances Immune Responses and Induces Regression of HER2 DCIS Independent of Route: Results of Randomized Selection Design Trial. Clin Cancer Res. 2017; 23(12): 2961–2971.
  84. Obara W, Kanehira M, Katagiri T, et al. Present status and future perspective of peptide-based vaccine therapy for urological cancer. Cancer Sci. 2018; 109(3): 550–559.
  85. Chamani R, Ranji P, Hadji M, et al. Application of E75 peptide vaccine in breast cancer patients: A systematic review and meta-analysis. Eur J Pharmacol. 2018; 831: 87–93.
  86. Hilf N, Kuttruff-Coqui S, Frenzel K, et al. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature. 2019; 565(7738): 240–245.
  87. Schirrmacher V, van Gool S, Stuecker W. Breaking Therapy Resistance: An Update on Oncolytic Newcastle Disease Virus for Improvements of Cancer Therapy. Biomedicines. 2019; 7(3).
  88. Artusio E, Hathaway B, Stanson J, et al. Transfection of human monocyte-derived dendritic cells with native tumor DNA induces antigen-specific T-cell responses in vitro. Cancer Biol Ther. 2006; 5(12): 1624–1631.
  89. Anguille S, Van de Velde AL, Smits EL, et al. Dendritic cell vaccination as postremission treatment to prevent or delay relapse in acute myeloid leukemia. Blood. 2017; 130(15): 1713–1721.
  90. Wang B, He J, Liu C, et al. An effective cancer vaccine modality: lentiviral modification of dendritic cells expressing multiple cancer-specific antigens. Vaccine. 2006; 24(17): 3477–3489.
  91. Van Gool SW, Makalowski J, Feyen O, et al. The induction of immunogenic cell death (ICD) during maintenance chemotherapy and subsequent multimudal immunotherapy for glioblastoma (GBM). Austin Oncol Case Rep. 2018; 3: 1010.
  92. Gubin MM, Artyomov MN, Mardis ER, et al. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest. 2015; 125(9): 3413–3421.
  93. Schirrmacher V. Oncolytic Newcastle disease virus as a prospective anti-cancer therapy. A biologic agent with potential to break therapy resistance. Expert Opin Biol Ther. 2015; 15(12): 1757–1771.
  94. Czarnywojtek A, Warmuz-Stangierska I, Woliński K, et al. Radioiodine therapy in patients with type II amiodarone-induced thyrotoxicosis. Pol Arch Med Wewn. 2014; 124(12): 695–703.
  95. Hermida J. Radioiodine ablation of the thyroid to prevent recurrence of amiodarone-induced thyrotoxicosis in patients with resistant tachyarrhythmias. Europace. 2004; 6(2): 169–174.
  96. Gursoy A, Tutuncu NB, Gencoglu A, et al. Radioactive iodine in the treatment of type-2 amiodarone-induced thyrotoxicosis. J Natl Med Assoc. 2008; 100(6): 716–719.
  97. Czarnywojtek A, Zgorzalewicz-Stachowiak M, Woliński K, et al. Results of preventive radioiodine therapy in euthyroid patients with history of hyperthyroidism prior to administration of amiodarone with permanent atrial fibrillation--a preliminary study. Endokrynol Pol. 2014; 65(4): 269–274.
  98. Joseph F, Younis N, Bowen‐Jones D. Treatment of carbimazole-induced agranulocytosis and sepsis with granulocyte colony stimulating factor. International Journal of Clinical Practice. 2003; 57(2): 145–146.
  99. Wiersinga WM. Amiodarone and the thyroid. In: Weetman AP, Grossman A. ed. Pharmacoterapeutics of the thyroid gland. Springer Verlag, Berlin 1997.
  100. Otsuka F, Noh JY, Chino T, et al. Hepatotoxicity and cutaneous reactions after antithyroid drug administration. Clin Endocrinol (Oxf). 2012; 77(2): 310–315.
  101. Faivre J, Clerc J, Gérolami R, et al. Long-term radioiodine retention and regression of liver cancer after sodium iodide symporter gene transfer in wistar rats. Cancer Res. 2004; 64(21): 8045–8051.
  102. Dwyer RM, Bergert ER, O'Connor MK, et al. Sodium iodide symporter-mediated radioiodide imaging and therapy of ovarian tumor xenografts in mice. Gene Ther. 2006; 13(1): 60–66.
  103. Mitrofanova E, Unfer R, Vahanian N, et al. Rat sodium iodide symporter for radioiodide therapy of cancer. Clin Cancer Res. 2004; 10(20): 6969–6976.
  104. Schipper ML, Weber A, Behe M, et al. Radioiodine treatment after sodium iodide symporter gene transfer is a highly effective therapy in neuroendocrine tumor cells. Cancer Res. 2003; 63(3): 1333–1338.
  105. Boland A, Ricard M, Opolon P, et al. Adenovirus-mediated transfer of the thyroid sodium/iodide symporter gene into tumors for a targeted radiotherapy. Cancer Res. 2000; 60(13): 3484–3492.
  106. Wolny M, Syrenicz A. [Sodium iodide symporter in physiology and diseases -- the current state of knowledge]. Endokrynol Pol. 2007; 58(6): 512–521.
  107. Spitzweg C, Joba W, Schriever K, et al. Analysis of human sodium iodide symporter immunoreactivity in human exocrine glands. J Clin Endocrinol Metab. 1999; 84(11): 4178–4184.
  108. Cho JY. Hormonal Regulation of Radioiodide Uptake Activity and Na+/I- Symporter Expression in Mammary Glands. J Clin Endocrinol Metab. 2000; 85(8): 2936–2943.
  109. Bruno R, Giannasio P, Ronga G, et al. Sodium iodide symporter expression and radioiodine distribution in extrathyroidal tissues. J Endocrinol Invest. 2004; 27(11): 1010–1014.
  110. Morgenstern KE, Vadysirisack DD, Zhang Z, et al. Expression of sodium iodide symporter in the lacrimal drainage system: implication for the mechanism underlying nasolacrimal duct obstruction in I(131)-treated patients. Ophthalmic Plast Reconstr Surg. 2005; 21(5): 337–344.
  111. Tazebay UH, Wapnir IL, Levy O, et al. The mammary gland iodide transporter is expressed during lactation and in breast cancer. Nat Med. 2000; 6(8): 871–878.
  112. Upadhyay G, Singh R, Agarwal G, et al. Functional expression of sodium iodide symporter (NIS) in human breast cancer tissue. Breast Cancer Res Treat. 2003; 77(2): 157–165.
  113. Smanik PA, Ryu KY, Theil KS, et al. Expression, exon-intron organization, and chromosome mapping of the human sodium iodide symporter. Endocrinology. 1997; 138(8): 3555–3558.
  114. Spitzweg C, Scholz IV, Bergert ER, et al. Retinoic acid-induced stimulation of sodium iodide symporter expression and cytotoxicity of radioiodine in prostate cancer cells. Endocrinology. 2003; 144(8): 3423–3432.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl