To the content
3 . 2023

The evolution of type 1 diabetes mellitus care

Abstract

Diabetes mellitus is currently a common disease, which is characterized by a high level of disability and mortality from complications. Nowadays in Russia there are about 229 thousand people among the adult population suffer from type 1 diabetes mellitus (T1DM), and about 48 thousand people among children and adolescents. To reduce the risk of developing diabetic complications, it is necessary to achieve compensation for diabetes and maintain blood glucose levels in the target range. However, this is not an easy task. Treatment of T1DM significantly reduces the quality of life, so researchers from different countries are searching for new ways of T1DM treatment that could maintain optimal control of carbohydrate metabolism, while not reducing the quality of life of patients and not limiting them. Since the 1920s, the main treatment for T1DM has been insulin administration. Since then, insulin production technologies have improved. In 1946 NPH-insulin was obtained, and in the early 2000s insulin analogues (glargine and detemir) were produced. Also, an important aspect of treatment is the optimization of the method of insulin administration. The first insulin pen appeared in 1981. A smart Bluetooth-enabled insulin pen that records the amount and time of insulin administration, displays the last dose, and also transfers information to a special mobile application appeared in 2017. A real breakthrough in the treatment of T1DM was the development of an insulin pump – ​a programmable device that continuously delivers rapid-acting insulin. Such a device helps patients to achieve diabetes compensation and improve the quality of life.

A promising and fundamentally new method of treating T1DM is transplantation of the entire pancreas or part of it. However, in this case, lifelong immunosuppression is required, and there is also the problem of the availability of islet donors and cell survival after the procedure. The possibility of using stem cells as a source of α- and β-cells and growing islets in vitro is being considered. At the same time, 3D bioprinting technology will ensure the correct distribution of cells in the scaffold, which will help solve the problems of creating a natural environment for pancreatic islets with an extracellular matrix and a vascular network. Currently, work is underway to improve this technology.

Keywords:type 1 diabetes mellitus; insulin therapy; syringe pen; insulin pump; self-control; transplantation; stem cells

Funding. The study had no sponsor support.

Conflict of interest. The authors declare no conflict of interest.

For citation: Bulgakova S.V., Dolgikh Yu.A., Sharonova L.A., Kosareva O.V., Treneva E.V., Kurmaev D.P., Lebedeva E.A., Galkin R.A. The evolution of type 1 diabetes care. Endokrinologiya: novosti, mneniya, obuchenie [Endocrinology: News, Opinions, Training]. 2023; 12 (3): 46–53. DOI: https://doi.org/10.33029/2304-9529-2023-12-3-46-53 (in Russian)

References

1. Plaksin N.S., Kupriyanova V.M., Bogdanova T.M. Diabetes mellitus: history of discovery, complications, prevalence. Mezhdunarodniy studencheskiy nauchniy vestnik [International Student Scientific Bulletin]. 2018; (5). URL: https://eduherald.ru/ru/article/view?id=19223 (date of access May 05, 2023). (in Russian)

2. URL: https://sd.diaregistry.ru/

3. Jabbour G., Henderson M., Mathieu M.E. Barriers to active lifestyles in children with type 1 diabetes. Can J Diabetes. 2016; 40 (2): 170–2.

4. Van Name M.A., Hilliard M.E., Boyle C.T., et al. Nighttime is the worst time: parental fear of hypoglycemia in young children with type 1 diabetes. Pediatr Diabetes. 2018; 19 (1): 114–20.

5. Foster N.C., Beck R.W., Miller K.M., et al. State of type 1 diabetes management and outcomes from the T1D exchange in 2016–2018. Diabetes Technol Ther. 2019; 21 (2): 66–72.

6. Pettus J.H., Zhou F.L., Shepherd L., et al. Incidences of severe hypoglycemia and diabetic ketoacidosis and prevalence of microvascular complications stratified by age and glycemic control in U.S. Adult patients with type 1 diabetes: a real-world study. Diabetes Care. 2019; 42 (12): 2220–7.

7. Ametov A.S., Pugovkina Ya.V., Vovk P.S. Insulin therapy is a century-long success story. Focus on basal insulin. Endokrinologiya: novosti, mneniya, obuchenie [Endocrinology: News, Opinions, Training]. 2021; 10 (1): 26–33. (in Russian)

8. Goeddel D.V., Kleid D.G., Bolivar F., Heyneker H.L., Yansura D.G., Crea R., et al. Expression in Escherichia coli of chemically synthesized genes for human insulin. Proc Natl Acad Sci USA. 1979; 76: 106–10.

9. Miller W.L., Baxter J.D. Recombinant DNA – a new source of insulin. Diabetologia. 1980; 18: 431–6.

10. Cereghino G.P.L., Cregg J.M. Applications of yeast in biotechnology: protein production and genetic analysis. Curr Opin Biotechnol. 1999; 10: 422–7.

11. Okminyan G.F., Latyshev O. Yu., Kiseleva E.V., Kasatkina E.P., Samsonova L.N. The evolution of basal insulin preparations: from simple to complex. Endokrinologiya: novosti, mneniya, obuchenie [Endocrinology: News, Opinions, Training]. 2021; 10 (1): 18–25. (in Russian)

12. Hirsch I.B., Juneja R., Beals J.M., Antalis C.J., Wright E.E. The Evolution of insulin and how it informs therapy and treatment choices // Endocr. Rev. 2020. Vol. 41, N 5. P. 733–755.

13. Pettus J., Santos Cavaiola T., Tamborlane W.V., Edelman S. The past, present, and future of basal insulins. Diabetes Metab Res Rev. 2016; 32 (6): 478–96.

14. Home P. The evolution of insulin therapy. Diabetes Res Clin Pract. 2021; 175: 108816.

15. Klonoff D.C., Aimbe F., Kerr D. Smart pens will improve insulin therapy. J Diabetes Sci Technol. 2018; 12 (3): 551–3.

16. Gomez-Peralta F., Abreu C., Gomez-Rodriguez S., et al. A novel insulin delivery optimization and tracking system. Diabetes Technol Ther. 2019; 21 (4): 209–14.

17. Laffel L.M., Limbert C., Phelan H., et al. ISPAD clinical practice consensus guidelines 2018: sick day management in children and adolescents with diabetes. Pediatr Diabetes. 2018; 19 (suppl 27): 193–204.

18. Zaharieva D.P., McGaugh S., Pooni R., Vienneau T., Ly T., Riddell M.C. Improved open-loop glucose control with basal insulin reduction 90 minutes before aerobic exercise in patients with type 1 diabetes on continuous subcutaneous insulin infusion. Diabetes Care. 2019; 42 (5): 824–31.

19. Pozzilli P., Battelino T., Danne T., et al. Continuous subcutaneous insulin infusion in diabetes: patient populations, safety, efficacy, and pharmacoeconomics. Diabetes Metab Res Rev. 2016; 32 (1): 21–39.

20. Misso M.L., Egberts K.J., Page M., O’Connor D., Shaw J. Continuous subcutaneous insulin infusion (CSII) versus multiple insulin injections for type 1 diabetes mellitus. Cochrane Database Syst Rev. 2010; 20 (1): CD 005103.

21. Karges B., Schwandt A., Heidtmann B., et al. Association of insulin pump therapy vs insulin injection therapy with severe hypoglycemia, ketoacidosis, and glycemic control among children, adolescents, and young adults with type 1 diabetes. JAMA. 2017; 318 (14): 1358–66.

22. Bollyky J.B., Bravata D., Yang J., Williamson M., Schneider J. Remote lifestyle coaching plus a connected glucose meter with certified diabetes educator support improves glucose and weight loss for people with type 2 diabetes. J Diabetes Res. 2018; 2018: 3961730.

23. Christiansen M., Greene C., Pardo S., et al. A new, wireless-enabled blood glucose monitoring system that links to a smart mobile device: accuracy and user performance evaluation. J Diabetes Sci Technol. 2017; 11 (3): 567–73.

24. Deiss D., Bolinder J., Riveline J.P., et al. Improved glycemic control in poorly controlled patients with type 1 diabetes using real-time continuous glucose monitoring. Diabetes Care. 2009; 29 (12): 2730–2.

25. Langendam M., Luijf Y.M., Hooft L., Devries J.H., Mudde A.H., Scholten R.J. Continuous glucose monitoring systems for type 1 diabetes mellitus. Cochrane Database Syst Rev. 2012; 1 (1): CD 008101.

26. Piona C., Dovc K., Mutlu G.Y., et al. Non-adjunctive flash glucose monitoring system use during summer-camp in children with type 1 diabetes: the free-summer study. Pediatr Diabetes. 2018; 19 (7): 1285–93.

27. Deiss D., Irace C., Carlson G., Tweden K.S., Kaufman F.R. Real-world safety of an implantable continuous glucose sensor over multiple cycles of use: a post-market registry study. Diabetes Technol Ther. 2020; 22 (1): 48–52.

28. Zaharieva D.P., Turksoy K., McGaugh S.M., et al. Lag time remains with newer real-time continuous glucose monitoring technology during aerobic exercise in adults living with type 1 diabetes. Diabetes Technol Ther. 2019; 21 (6): 313–21.

29. DeSalvo D.J., Miller K.M., Hermann J.M., et al. Continuous glucose monitoring and glycemic control among youth with type 1 diabetes: international comparison from the T1D Exchange and DPV Initiative. Pediatr Diabetes. 2018; 19 (7): 1271–5.

30. Dovc K., Cargnelutti K., Sturm A., Selb J., Bratina N., Battelino T. Continuous glucose monitoring use and glucose variability in pre-school children with type 1 diabetes. Diabetes Res Clin Pract. 2019; 147: 76–80.

31. Bergenstal R.M. Continuous glucose monitoring: transforming diabetes management step by step. Lancet. 2018; 391 (10 128): 1334–6.

32. Kropff J., Choudhary P., Neupane S., et al. Accuracy and longevity of an implantable continuous glucose sensor in the PRECISE study: a 180-day, prospective, multicenter, pivotal trial. Diabetes Care. 2017; 40 (1): 63–8.

33. Polonsky W.H., Hessler D., Ruedy K.J., et al. The impact of continuous glucose monitoring on markers of quality of life in adults with type 1 diabetes: further findings from the DIAMOND randomized clinical trial. Diabetes Care. 2017; 40 (6): 736–41.

34. Nimri R., Ochs A.R., Pinsker J.E., et al. Decision support systems and closed loop. Diabetes Technol Ther. 2019; 21 (S 1): S 42–56.

35. Shapiro A.M.J., Lakey J.R.T., Ryan E.A., et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000; 343 (4): 230–8.

36. Docherty F.M., Sussel L. Islet regeneration: endogenous and exogenous approaches. Int J Mol Sci. 2021; 22 (7): 3306.

37. Yang J., Liu H., Sun H., Wang Z., Zhang R., Liu Y., et al. construction of induced pluripotent stem cell line (ZZUi0017-A) from the fibroblast cells of a female patient with CACNA1A mutation by unintegrated reprogramming approach. Stem Cell Res. 2020; 48: 101946.

38. Wobus A.M., Boheler K.R. Embryonic stem cells: prospects for developmental biology and cell therapy. Physiol Rev. 2005; 85 (2): 635–78.

39. Klimanskaya I., Chung Y., Becker S., Lu S.J., Lanza R. Human embryonic stem cell lines derived from single blastomeres. Nature. 2006; 444 (7118): 481–5.

40. Baylis F. Human embryonic stem cell lines: the ethics of derivation. J Obstet Gynaecol Can. 2002; 24 (2): 159–63.

41. Hovatta O., Stojkovic M., Nogueira M., Varela-Nieto I. European scientific, ethical, and legal issues on human stem cell research and regenerative medicine. Stem Cells. 2010; 28 (6): 1005–7.

42. Friedenstein A.J., Chailakhyan R.K., Latsinik N.V., Panasyvk A.F., Keiliss-Borok I.V. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues: cloning in vitro and retransplantation in vivo. Transplantation. 1974; 17 (4): 331–40.

43. Sousa B.R., Parreira R.C., Fonseca E.A., Amaya M.J., Tonelli F.M.P., Lacerda S.M.S.N., et al. Human adult stem cells from diverse origins: an overview from multiparametric immunophenotyping to clinical applications. Cytometry A. 2014; 85 (1): 43–77.

44. Zuk P.A., Zhu M., Mizuno H., Huang J., Futrell J.W., Katz A.J., et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001; 7 (2): 211–28.

45. Prabakar K.R., Domínguez-Bendala J., Damaris Molano R., Pileggi A., Villate S., Ricordi C., et al. Generation of glucose-responsive, insulin-producing cells from human umbilical cord blood-derived mesenchymal stem cells. Cell Transplant. 2012; 21 (6): 1321–39.

46. Shivakumar S.B., Lee H.J., Son Y.B., Bharti D., Ock S.A., Lee S.L., et al. In vitro differentiation of single donor derived human dental mesenchymal stem cells into pancreatic β cell-like cells. Biosci Rep. 2019; 39 (5): BSR 20182051.

47. Kanafi M.M., Rajeshwari Y.B., Gupta S., Dadheech N., Nair P.D., Gupta P.K., et al. Transplantation of islet-like cell clusters derived from human dental pulp stem cells restores normoglycemia in diabetic mice. Cytotherapy. 2013; 15 (10): 1228–36.

48. Guo Q.S., Zhu M.Y., Wang L., Fan X.J., Lu Y.H., Wang Z.W., et al. Combined transfection of the three transcriptional factors, PDX-1, NeuroD 1, and MafA, causes differentiation of bone marrow mesenchymal stem cells into insulin-producing cells. Exp Diabetes Res. 2012; 2012: 672013.

49. Lechner A., Yang Y.-G., Blacken R.A., Wang L., Nolan A.L., Habener J.F. No evidence for significant transdifferentiation of bone marrow into pancreatic-cells in vivo. Diabetes. 2004; 53 (3): 616–23.

50. Choi J.B., Uchino H., Azuma K., Iwashita N., Tanaka Y., Mochizuki H., et al. Little evidence of transdifferentiation of bone marrow-derived cells into pancreatic beta cells. Diabetologia. 2003; 46 (10): 1366–74.

51. Ezquer F., Ezquer M., Contador D., Ricca M., Simon V., Conget P. The antidiabetic effect of mesenchymal stem cells is unrelated to their transdifferentiation potential but to their capability to restore Th1/Th2 balance and to modify the pancreatic microenvironment. Stem Cells. 2012; 30 (8): 1664–74.

52. Dave S.D., Vanikar A.V., Trivedi H.L., Thakkar U.G., Gopal S.C., Chandra T. Novel therapy for insulin-dependent diabetes mellitus: infusion of in vitro-generated insulin-secreting cells. Clin Exp Med. 2015; 15 (1): 41–5.

53. Thakkar U.G., Trivedi H.L., Vanikar A.V., Dave S.D. Insulin-secreting adipose-derived mesenchymal stromal cells with bone marrow-derived hematopoietic stem cells from autologous and allogenic sources for type 1 diabetes mellitus. Cytotherapy. 2015; 17 (7): 940–7.

54. Intravenous infusion of human adipose tissue-derived mesenchymal stem cells to treat type 1 diabetic mellitus in mice: an evaluation of grafted cell doses. Adv Exp Med Biol. 2018; 1083: 145–56.

55. Li L., Hui H., Jia X., Zhang J., Liu Y., Xu Q., Zhu D. Infusion with human bone marrow-derived mesenchymal stem cells improves β-cell function in patients and non-obese mice with severe diabetes. Sci Rep. 2016; 6: 37894.

56. Yaochite J.N.U., Caliari-Oliveira C., de Souza L.E.B., Neto L.S., Palma P.V.B., Covas D.T., et al. Therapeutic efficacy and biodistribution of allogeneic mesenchymal stem cells delivered by intrasplenic and intrapancreatic routes in streptozotocin-induced diabetic mice. Stem Cell Res Ther. 2015; 6 (1): 31.

57. Chen J., Chen J., Cheng Y., Fu Y., Zhao H., Tang M., et al. Mesenchymal stem cell-derived exosomes protect beta cells against hypoxia-induced apoptosis via MiR-21 by alleviating ER stress and inhibiting P38 MAPK phosphorylation. Stem Cell Res Ther. 2020; 11 (1): 97.

58. Mesples A., Majeed N., Zhang Y., Xiang H. Early immunotherapy using autologous adult stem cells reversed the effect of anti-pancreatic islets in recently diagnosed type 1 diabetes mellitus: preliminary results. Med Sci Monit. 2013; 19: 852–7.

59. Carlsson P.O., Schwarcz E., Korsgren O., le Blanc K. Preserved β-cell function in type 1 diabetes by mesenchymal stromal cells. Diabetes. 2015; 64 (2): 587–92.

60. Takahashi K., Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006; 126 (4): 663–76.

61. Takahashi K., Tanabe K., Ohnuki M., Narita M., Ichisaka T., Tomoda K., Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007; 131 (5): 861–72.

62. Okit K., Yamakawa T., Matsumura Y., Sato Y., Amano N., Watanabe A., et al. An efficient nonviral method to generate integration-free human-induced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells. 2013; 31 (3): 458–66.

63. Xue Y., Cai X., Wang L., Liao B., Zhang H., Shan Y., et al. Generating a non-integrating human induced pluripotent stem cell bank from urine-derived cells. PLoS One. 2013; 8 (8): e70573.

64. Moede T., Leibiger I.B., Berggren P.O. Alpha cell regulation of beta cell function. Diabetologia. 2020; 63 (10): 2064–75.

65. Adams M.T., Gilbert J.M., Hinojosa Paiz J., Bowman F.M., Blum B. Endocrine cell type sorting and mature architecture in the islets of langerhans require expression of roundabout receptors in β cells. Sci Rep. 2018; 8 (1): 10876.

66. Caicedo A. Paracrine and autocrine interactions in the human islet: more than meets the eye. Semin Cell Dev Biol. 2013; 24 (1): 11–21.

67. URL: https://habr.com/ru/post/445020/

All articles in our journal are distributed under the Creative Commons Attribution 4.0 International License (CC BY 4.0 license)

CHIEF EDITOR
CHIEF EDITOR
Ametov Alexander S.
Honored Scientist of the Russian Federation, Doctor of Medical Sciences, Professor, Head of Subdepartment of Endocrinology, Head of the UNESCO Network Chair on the subject «Bioethics of diabetes as a global problem» of the Russian Medical Academy of Continuous Professional Education (Moscow)
Вскрытие

Journals of «GEOTAR-Media»