open access

Vol 57, No 1 (2019)
Review paper
Submitted: 2019-02-05
Accepted: 2019-02-06
Published online: 2019-03-11
Get Citation

In vitro generation of pancreatic β-cells for diabetes treatment. I. β-like cells derived from human pluripotent stem cells

Katarzyna Cierpka-Kmiec1, Agata Wronska2, Zbigniew Kmiec2
·
Pubmed: 30869153
·
Folia Histochem Cytobiol 2019;57(1):1-14.
Affiliations
  1. Department of Hypertension and Diabetology, Medical University of Gdansk, Gdansk, Poland
  2. Department of Histology, Medical University of Gdansk, Poland

open access

Vol 57, No 1 (2019)
REVIEW
Submitted: 2019-02-05
Accepted: 2019-02-06
Published online: 2019-03-11

Abstract

Diabetes mellitus is a chronic disease that affects hundreds of millions of people worldwide. Type 1 diabetes (T1D) is characterized by the lack of pancreatic β-cells that had been destroyed as a result of an autoimmune response. Therefore, in patients with T1D, the replacement therapy with functional β-cells derived from extrinsic sources could be a preferable option as compared to insulin treatment. Unfortunately, successful transplantation of whole pancreata or pancreatic islets into patients with diabetes is available only to a fraction of them due to the scarcity of donors. The rapid development of cell reprogramming methods made it possible to generate large numbers of human β-like cells derived from human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs). This review describes the basis of in vitro differentiaton protocols of β-like cells that mimic changes of the main signaling pathways during the key stages of human and murine pancreas development, which are described first. During the last 15 years it was found that there are no important differences between hESCs and hiPSCs in their differentiation capacities into β-like cells and the expression profiles of the key transcription factors. The in vitro produced β-like cells are immature as demonstrated by functional tests in rodents and single-cell transcriptomic and proteomic analyses. After the transplantation of the β cell progenitors into immunocompromised diabetic mice, a few weeks have to pass before the increased insulin levels in response to glucose load appear. There is a continuous progress in the development of open-type encapsulation devices which allow the vascularization of the transplanted cells and protect them against host’s immune cells. The results of the first clinical trial of human partially differentiated endocrine progenitors of β cells transplanted into patients with T1D will be published in the year 2019. It is hoped that further improvements in the techniques of large-scale generation of the β-like cells derived from human pluripotent stem cells will bring us closer to their clinical application as a form of cause-directed therapy for people with diabetes.

 

Abstract

Diabetes mellitus is a chronic disease that affects hundreds of millions of people worldwide. Type 1 diabetes (T1D) is characterized by the lack of pancreatic β-cells that had been destroyed as a result of an autoimmune response. Therefore, in patients with T1D, the replacement therapy with functional β-cells derived from extrinsic sources could be a preferable option as compared to insulin treatment. Unfortunately, successful transplantation of whole pancreata or pancreatic islets into patients with diabetes is available only to a fraction of them due to the scarcity of donors. The rapid development of cell reprogramming methods made it possible to generate large numbers of human β-like cells derived from human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs). This review describes the basis of in vitro differentiaton protocols of β-like cells that mimic changes of the main signaling pathways during the key stages of human and murine pancreas development, which are described first. During the last 15 years it was found that there are no important differences between hESCs and hiPSCs in their differentiation capacities into β-like cells and the expression profiles of the key transcription factors. The in vitro produced β-like cells are immature as demonstrated by functional tests in rodents and single-cell transcriptomic and proteomic analyses. After the transplantation of the β cell progenitors into immunocompromised diabetic mice, a few weeks have to pass before the increased insulin levels in response to glucose load appear. There is a continuous progress in the development of open-type encapsulation devices which allow the vascularization of the transplanted cells and protect them against host’s immune cells. The results of the first clinical trial of human partially differentiated endocrine progenitors of β cells transplanted into patients with T1D will be published in the year 2019. It is hoped that further improvements in the techniques of large-scale generation of the β-like cells derived from human pluripotent stem cells will bring us closer to their clinical application as a form of cause-directed therapy for people with diabetes.

 

Get Citation

Keywords

diabetes; human pancreas development; hESC; hiPSC; in vitro differentiation; transcription factors; β-like cells; β cell replacement

About this article
Title

In vitro generation of pancreatic β-cells for diabetes treatment. I. β-like cells derived from human pluripotent stem cells

Journal

Folia Histochemica et Cytobiologica

Issue

Vol 57, No 1 (2019)

Article type

Review paper

Pages

1-14

Published online

2019-03-11

Page views

5408

Article views/downloads

3484

DOI

10.5603/FHC.a2019.0001

Pubmed

30869153

Bibliographic record

Folia Histochem Cytobiol 2019;57(1):1-14.

Keywords

diabetes
human pancreas development
hESC
hiPSC
in vitro differentiation
transcription factors
β-like cells
β cell replacement

Authors

Katarzyna Cierpka-Kmiec
Agata Wronska
Zbigniew Kmiec

References (97)
  1. Ogurtsova K, da Rocha Fernandes JD, Huang Y, et al. IDF Diabetes Atlas: Global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes Res Clin Pract. 2017; 128: 40–50.
  2. Gu G, Brown J, Melton D. Direct lineage tracing reveals the ontogeny of pancreatic cell fates during mouse embryogenesis. Mechanisms of Development. 2003; 120(1): 35–43.
  3. Pictet RL, Clark WR, Williams RH, et al. An ultrastructural analysis of the developing embryonic pancreas. Dev Biol. 1972; 29(4): 436–467.
  4. Slack JMW: Developmental biology of the pancreas. Development 1995;121:1569–1580, indexed in Pubmed: 7600975. .
  5. Pan FC, Wright C. Pancreas organogenesis: from bud to plexus to gland. Dev Dyn. 2011; 240(3): 530–565.
  6. Jørgensen MC, Ahnfelt-Rønne J, Hald J, et al. An illustrated review of early pancreas development in the mouse. Endocr Rev. 2007; 28(6): 685–705.
  7. Villasenor A, Chong DC, Cleaver O. Biphasic Ngn3 expression in the developing pancreas. Dev Dyn. 2008; 237(11): 3270–3279.
  8. Herrera PL. Adult insulin- and glucagon-producing cells differentiate from two independent cell lineages. Development. 2000; 127(11): 2317–2322.
  9. Bankaitis ED, Bechard ME, Wright CVE. Feedback control of growth, differentiation, and morphogenesis of pancreatic endocrine progenitors in an epithelial plexus niche. Genes Dev. 2015; 29(20): 2203–2216.
  10. Kesavan G, Sand FW, Greiner TU, et al. Cdc42-mediated tubulogenesis controls cell specification. Cell. 2009; 139(4): 791–801.
  11. Larsen HL, Grapin-Botton A. The molecular and morphogenetic basis of pancreas organogenesis. Semin Cell Dev Biol. 2017; 66: 51–68.
  12. Gradwohl G, Dierich A, LeMeur M, et al. Neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc Natl Acad Sci U S A. 2000; 97(4): 1607–1611.
  13. Gu G, Dubauskaite J, Melton DA. Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development. 2002; 129(10): 2447–2457.
  14. Desgraz R, Herrera PL. Pancreatic neurogenin 3-expressing cells are unipotent islet precursors. Development. 2009; 136(21): 3567–3574.
  15. Jennings RE, Berry AA, Kirkwood-Wilson R, et al. Development of the human pancreas from foregut to endocrine commitment. Diabetes. 2013; 62(10): 3514–3522.
  16. Pan FC, Brissova M. Pancreas development in humans. Curr Opin Endocrinol Diabetes Obes. 2014; 21(2): 77–82.
  17. Jennings RE, Berry AA, Strutt JP, et al. Human pancreas development. Development. 2015; 142(18): 3126–3137.
  18. Shih HP, Wang A, Sander M. Pancreas organogenesis: from lineage determination to morphogenesis. Annu Rev Cell Dev Biol. 2013; 29: 81–105.
  19. Al-Khawaga S, Memon B, Butler AE, et al. Pathways governing development of stem cell-derived pancreatic β cells: lessons from embryogenesis. Biol Rev Camb Philos Soc. 2018; 93(1): 364–389.
  20. Piper K, Brickwood S, Turnpenny LW, et al. Beta cell differentiation during early human pancreas development. J Endocrinol. 2004; 181(1): 11–23.
  21. Jeon J, Correa-Medina M, Ricordi C, et al. Endocrine cell clustering during human pancreas development. J Histochem Cytochem. 2009; 57(9): 811–824.
  22. Lyttle BM, Li J, Krishnamurthy M, et al. Transcription factor expression in the developing human fetal endocrine pancreas. Diabetologia. 2008; 51(7): 1169–1180.
  23. Riedel MJ, Asadi A, Wang R, et al. Immunohistochemical characterisation of cells co-producing insulin and glucagon in the developing human pancreas. Diabetologia. 2012; 55(2): 372–381.
  24. Dassaye R, Naidoo S, Cerf ME. Transcription factor regulation of pancreatic organogenesis, differentiation and maturation. Islets. 2016; 8(1): 13–34.
  25. Conrad E, Stein R, Hunter CS. Revealing transcription factors during human pancreatic β cell development. Trends Endocrinol Metab. 2014; 25(8): 407–414.
  26. Schaffer AE, Freude KK, Nelson SB, et al. Nkx6 transcription factors and Ptf1a function as antagonistic lineage determinants in multipotent pancreatic progenitors. Dev Cell. 2010; 18(6): 1022–1029.
  27. Offield MF, Jetton TL, Labosky PA, et al. PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development. 1996; 122(3): 983–995.
  28. Stoffers DA, Zinkin NT, Stanojevic V, et al. Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat Genet. 1997; 15(1): 106–110.
  29. Wang X, Sterr M, Burtscher I, et al. Genome-wide analysis of PDX1 target genes in human pancreatic progenitors. Mol Metab. 2018; 9: 57–68.
  30. Sellick GS, Barker KT, Stolte-Dijkstra I, et al. Mutations in PTF1A cause pancreatic and cerebellar agenesis. Nat Genet. 2004; 36(12): 1301–1305.
  31. Reichert M, Rustgi AK. Pancreatic ductal cells in development, regeneration, and neoplasia. J Clin Invest. 2011; 121(12): 4572–4578.
  32. Benitez CM, Goodyer WR, Kim SK. Deconstructing pancreas developmental biology. Cold Spring Harb Perspect Biol. 2012; 4(6).
  33. Johansson KA, Dursun U, Jordan N, et al. Temporal control of neurogenin3 activity in pancreas progenitors reveals competence windows for the generation of different endocrine cell types. Dev Cell. 2007; 12(3): 457–465.
  34. Salisbury RJ, Blaylock J, Berry AA, et al. The window period of NEUROGENIN3 during human gestation. Islets. 2014; 6(3): e954436.
  35. Rubio-Cabezas O, Codner E, Flanagan SE, et al. Neurogenin 3 is important but not essential for pancreatic islet development in humans. Diabetologia. 2014; 57(11): 2421–2424.
  36. Zhu Y, Liu Q, Zhou Z, et al. PDX1, Neurogenin-3, and MAFA: critical transcription regulators for beta cell development and regeneration. Stem Cell Res Ther. 2017; 8(1): 240.
  37. Pinney S, Oliver-Krasinski J, Ernst L, et al. Neonatal Diabetes and Congenital Malabsorptive Diarrhea Attributable to a Novel Mutation in the Human Neurogenin-3 Gene Coding Sequence. The Journal of Clinical Endocrinology & Metabolism. 2011; 96(7): 1960–1965.
  38. Sander M, Sussel L, Conners J, et al. Homeobox gene Nkx6.1 lies downstream of Nkx2.2 in the major pathway of beta-cell formation in the pancreas. Development. 2000; 127(24): 5533–5540.
  39. Petersen MB, Azad A, Ingvorsen C, et al. Single-Cell Gene Expression Analysis of a Human ESC Model of Pancreatic Endocrine Development Reveals Different Paths to β-Cell Differentiation. Stem Cell Reports. 2017; 9(4): 1246–1261.
  40. Matsuoka Ta, Artner I, Henderson E, et al. The MafA transcription factor appears to be responsible for tissue-specific expression of insulin. Proc Natl Acad Sci U S A. 2004; 101(9): 2930–2933.
  41. Santosa MM, Low BS, Pek NM, et al. Knowledge Gaps in Rodent Pancreas Biology: Taking Human Pluripotent Stem Cell-Derived Pancreatic Beta Cells into Our Own Hands. Front Endocrinol (Lausanne). 2015; 6: 194.
  42. McCracken KW, Wells JM. Molecular pathways controlling pancreas induction. Semin Cell Dev Biol. 2012; 23(6): 656–662.
  43. Gittes GK. Developmental biology of the pancreas: a comprehensive review. Dev Biol. 2009; 326(1): 4–35.
  44. Nair G, Hebrok M. Islet formation in mice and men: lessons for the generation of functional insulin-producing β-cells from human pluripotent stem cells. Curr Opin Genet Dev. 2015; 32: 171–180.
  45. Balboa D, Saarimäki-Vire J, Otonkoski T. Concise Review: Human Pluripotent Stem Cells for the Modeling of Pancreatic β-Cell Pathology. Stem Cells. 2019; 37(1): 33–41.
  46. Wilson ME, Scheel D, German MS. Gene expression cascades in pancreatic development. Mech Dev. 2003; 120(1): 65–80.
  47. Jennings RE, Berry AA, Gerrard DT, et al. Laser Capture and Deep Sequencing Reveals the Transcriptomic Programmes Regulating the Onset of Pancreas and Liver Differentiation in Human Embryos. Stem Cell Reports. 2017; 9(5): 1387–1394.
  48. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998; 282(5391): 1145–1147.
  49. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–676. .
  50. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007; 131(5): 861–872.
  51. Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007; 318(5858): 1917–1920.
  52. Sisakhtnezhad S, Matin MM. Transdifferentiation: a cell and molecular reprogramming process. Cell Tissue Res. 2012; 348(3): 379–396.
  53. D'Amour KA, Agulnick AD, Eliazer S, et al. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol. 2005; 23(12): 1534–1541.
  54. D'Amour KA, Bang AG, Eliazer S, et al. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat Biotechnol. 2006; 24(11): 1392–1401.
  55. Kroon E, Martinson LA, Kadoya K, et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol. 2008; 26(4): 443–452.
  56. Basford CL, Prentice KJ, Hardy AB, et al. The functional and molecular characterisation of human embryonic stem cell-derived insulin-positive cells compared with adult pancreatic beta cells. Diabetologia. 2012; 55(2): 358–371.
  57. Fujikawa T, Oh SH, Pi L, et al. Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell-derived insulin-producing cells. Am J Pathol. 2005; 166(6): 1781–1791.
  58. Rezania A, Bruin JE, Arora P, et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. 2014; 32(11): 1121–1133.
  59. Pagliuca FW, Millman JR, Gürtler M, et al. Generation of functional human pancreatic β cells in vitro. Cell. 2014; 159(2): 428–439.
  60. Russ HA, Parent AV, Ringler JJ, et al. Controlled induction of human pancreatic progenitors produces functional beta-like cells in vitro. EMBO J. 2015; 34(13): 1759–1772.
  61. Yoshioka M, Kayo T, Ikeda T, et al. A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes. 1997; 46(5): 887–894.
  62. Ma X, Zhu S. Chemical strategies for pancreatic β cell differentiation, reprogramming, and regeneration. Acta Biochim Biophys Sin (Shanghai). 2017; 49(4): 289–301.
  63. Yabe SG, Fukuda S, Takeda F, et al. Efficient generation of functional pancreatic β-cells from human induced pluripotent stem cells. J Diabetes. 2017; 9(2): 168–179.
  64. Memon B, Karam M, Al-Khawaga S, et al. Enhanced differentiation of human pluripotent stem cells into pancreatic progenitors co-expressing PDX1 and NKX6.1. Stem Cell Res Ther. 2018; 9(1): 15.
  65. Ida H, Akiyama T, Ishiguro K, et al. Establishment of a rapid and footprint-free protocol for differentiation of human embryonic stem cells into pancreatic endocrine cells with synthetic mRNAs encoding transcription factors. Stem Cell Res Ther. 2018; 9(1): 277.
  66. Kondo Y, Toyoda T, Ito R, et al. Identification of a small molecule that facilitates the differentiation of human iPSCs/ESCs and mouse embryonic pancreatic explants into pancreatic endocrine cells. Diabetologia. 2017; 60(8): 1454–1466.
  67. Aghazadeh Y, Nostro MC. Cell Therapy for Type 1 Diabetes: Current and Future Strategies. Curr Diab Rep. 2017; 17(6): 37.
  68. Hashemitabar M, Heidari E. Redefining the signaling pathways from pluripotency to pancreas development: In vitro β-cell differentiation. J Cell Physiol. 2019; 234(6): 7811–7827.
  69. Millman JR, Pagliuca FW. Autologous Pluripotent Stem Cell-Derived β-Like Cells for Diabetes Cellular Therapy. Diabetes. 2017; 66(5): 1111–1120.
  70. Shahjalal HMd, Shiraki N, Sakano D, et al. Generation of insulin-producing β-like cells from human iPS cells in a defined and completely xeno-free culture system. J Mol Cell Biol. 2014; 6(5): 394–408.
  71. Singh SP, Ninov N. The triumvirate of beta-cell regeneration: solutions and bottlenecks to curing diabetes. Int J Dev Biol. 2018; 62(6-7-8): 453–464.
  72. Borowiak M, Maehr R, Chen S, et al. Small molecules efficiently direct endodermal differentiation of mouse and human embryonic stem cells. Cell Stem Cell. 2009; 4(4): 348–358.
  73. Ninomiya H, Mizuno K, Terada R, et al. Improved efficiency of definitive endoderm induction from human induced pluripotent stem cells in feeder and serum-free culture system. In Vitro Cell Dev Biol Anim. 2015; 51(1): 1–8.
  74. Chen S, Borowiak M, Fox JL, et al. A small molecule that directs differentiation of human ESCs into the pancreatic lineage. Nat Chem Biol. 2009; 5(4): 258–265.
  75. Trott J, Tan EeK, Ong S, et al. Long-Term Culture of Self-renewing Pancreatic Progenitors Derived from Human Pluripotent Stem Cells. Stem Cell Reports. 2017; 8(6): 1675–1688.
  76. Southard SM, Kotipatruni RP, Rust WL. Generation and selection of pluripotent stem cells for robust differentiation to insulin-secreting cells capable of reversing diabetes in rodents. PLoS One. 2018; 13(9): e0203126.
  77. Kelly OG, Chan MY, Martinson LA, et al. Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat Biotechnol. 2011; 29(8): 750–756.
  78. Jiang W, Sui X, Zhang D, et al. CD24: a novel surface marker for PDX1-positive pancreatic progenitors derived from human embryonic stem cells. Stem Cells. 2011; 29(4): 609–617.
  79. Ameri J, Borup R, Prawiro C, et al. Efficient Generation of Glucose-Responsive Beta Cells from Isolated GP2 Human Pancreatic Progenitors. Cell Rep. 2017; 19(1): 36–49.
  80. Cogger KF, Sinha A, Sarangi F, et al. Glycoprotein 2 is a specific cell surface marker of human pancreatic progenitors. Nat Commun. 2017; 8(1): 331.
  81. Ramond C, Glaser N, Berthault C, et al. Reconstructing human pancreatic differentiation by mapping specific cell populations during development. Elife. 2017; 6.
  82. Bonfanti P, Nobecourt E, Oshima M, et al. Ex Vivo Expansion and Differentiation of Human and Mouse Fetal Pancreatic Progenitors Are Modulated by Epidermal Growth Factor. Stem Cells Dev. 2015; 24(15): 1766–1778.
  83. Ramond C, Beydag-Tasöz BS, Azad A, et al. Understanding human fetal pancreas development using subpopulation sorting, RNA sequencing and single-cell profiling. Development. 2018; 145(16).
  84. Vieira A, Druelle N, Avolio F, et al. β-Cell Replacement Strategies: The Increasing Need for a. Front Genet. 2017; 8: 75.
  85. Kefaloyianni E, Lyssand JS, Moreno C, et al. Comparative proteomic analysis of the ATP-sensitive K+ channel complex in different tissue types. Proteomics. 2013; 13(2): 368–378.
  86. Osafune K, Caron L, Borowiak M, et al. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat Biotechnol. 2008; 26(3): 313–315.
  87. Maehr R, Chen S, Snitow M, et al. Generation of pluripotent stem cells from patients with type 1 diabetes. Proc Natl Acad Sci U S A. 2009; 106(37): 15768–15773.
  88. Thatava T, Kudva YC, Edukulla R, et al. Intrapatient variations in type 1 diabetes-specific iPS cell differentiation into insulin-producing cells. Mol Ther. 2013; 21(1): 228–239.
  89. Millman JR, Xie C, Van Dervort A, et al. Generation of stem cell-derived β-cells from patients with type 1 diabetes. Nat Commun. 2016; 7: 11463.
  90. Pellegrini S, Manenti F, Chimienti R, et al. Differentiation of Sendai Virus-Reprogrammed iPSC into β Cells, Compared with Human Pancreatic Islets and Immortalized β Cell Line. Cell Transplantation. 2018; 27(10): 1548–1560.
  91. Teo AKK, Windmueller R, Johansson BB, et al. Derivation of human induced pluripotent stem cells from patients with maturity onset diabetes of the young. J Biol Chem. 2013; 288(8): 5353–5356.
  92. Yabe SG, Iwasaki N, Yasuda K, et al. Establishment of maturity-onset diabetes of the young-induced pluripotent stem cells from a Japanese patient. J Diabetes Investig. 2015; 6(5): 543–547.
  93. Zhu S, Russ HA, Wang X, et al. Human pancreatic beta-like cells converted from fibroblasts. Nat Commun. 2016; 7: 10080.
  94. Vegas AJ, Veiseh O, Gürtler M, et al. Long-term glycemic control using polymer-encapsulated human stem cell-derived beta cells in immune-competent mice. Nat Med. 2016; 22(3): 306–311.
  95. Kasputis T, Clough D, Noto F, et al. Microporous Polymer Scaffolds for the Transplantation of Embryonic Stem Cell Derived Pancreatic Progenitors to a Clinically Translatable Site for the Treatment of Type I Diabetes. ACS Biomater Sci Eng. 2018; 4(5): 1770–1778.
  96. Rios PD, Skoumal M, Liu J, et al. Evaluation of encapsulating and microporous nondegradable hydrogel scaffold designs on islet engraftment in rodent models of diabetes. Biotechnol Bioeng. 2018; 115(9): 2356–2364.
  97. Schuetz C, Anazawa T, Cross SE, et al. IPITA YIC Young Investigator Committee. β Cell Replacement Therapy: The Next 10 Years. Transplantation. 2018; 102(2): 215–229.

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.

By VM Media Group sp z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

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