A preliminary evaluation of efficacy and safety of Wharton's jelly mesenchymal stem cell transplantation in patients with type 2 diabetes mellitus

Xuebin Liu, Pei Zheng, Xiaodong Wang, Guanghui Dai, Hongbin Cheng, Zan Zhang, Rongrong Hua, Xinxin Niu, Jing Shi, Yihua An, Xuebin Liu, Pei Zheng, Xiaodong Wang, Guanghui Dai, Hongbin Cheng, Zan Zhang, Rongrong Hua, Xinxin Niu, Jing Shi, Yihua An

Abstract

Introduction: Stem cell therapy has recently been introduced to treat patients with type 2 diabetes mellitus (T2DM). However, no data are available on the efficacy and safety of allogeneic Wharton's Jelly-derived mesenchymal stem cell (WJ-MSC) transplantation in patients with T2DM. Here we performed a non-placebo controlled prospective phase I/II study to determine efficacy and safety of WJ-MSC transplantation in T2DM.

Methods: Twenty-two patients with T2DM were enrolled and received WJ-MSC transplantation through one intravenous injection and one intrapancreatic endovascular injection (catheterization). They were followed up for 12 months after transplantation. The primary endpoints were changes in the levels of glycated hemoglobin and C-peptide and the secondary endpoints included insulin dosage, fasting blood glucose (FBG), post-meal blood glucose (PBG), inflammatory markers and T lymphocyte counts.

Results: WJ-MSC transplantation significantly decreased the levels of glucose and glycated hemoglobin, improved C-peptide levels and beta cell function, and reduced markers of systemic inflammation and T lymphocyte counts. No major WJ-MSC transplantation-related adverse events occurred, but data suggest a temporary decrease in levels of C-peptide and beta cell function at one month after treatment, possibly related to intrapancreatic endovascular injection.

Conclusions: Our data demonstrate that treatment with WJ-MSCs can improve metabolic control and beta cell function in patients with T2DM. The therapeutic mechanism may involve improvements in systemic inflammation and/or immunological regulation.

Trial registration: Chinese Clinical Trial Register ChiCTR-ONC-10000985. Registered 23 September 2010.

Figures

Figure 1
Figure 1
Fluorescence-activated cell sorting analysis of umbilical cord mesenchymal stem cells. Percentages of all CD105, CD73 and CD44 were higher than 95%, while none of CD34, CD45, CD31, CD146 and HLA-DR’s percentage was higher than 1%. WJ-MSCs, Wharton’s Jelly mesenchymal stem cells.
Figure 2
Figure 2
Changes in glycated hemoglobin, fasting glucose and 2 h postprandial glucose levels during the 12-month study period. A: The changes in glycated hemoglobin levels between baseline and 1, 3, 6 and 12 months. B: Changes in fasting glucose and 2 h postprandial glucose levels between baseline and 1, 3, 6 and 12 months. The results are shown as the mean ± S.D. *P <0.05 compared with pretreatment; **P <0.01 compared with pretreatment.
Figure 3
Figure 3
Changes in C-peptide levels and beta-cell secretory function during the 12-month study period. A: The changes in fasting C-peptide levels and OGTT 2 h postprandial C-peptide levels between baseline and 1, 3, 6 and 12 months. B: Beta-cell secretory function was assessed with HOMA-2B. The results are shown as the mean ± S.D. *P <0.05 compared with pretreatment. HOMA, homeostasis model assessment; OGTT, oral glucose tolerance test.
Figure 4
Figure 4
Changes in daily insulin requirements over time. This figure shows the insulin requirements of T2DM patients at pre-operation and 1, 3, 6 and 12 months post-transplantation. The results are shown as the mean ± S.D. *P <0.05 compared with pretreatment. The analysis included 17 patients that received exogenous insulin at months 0, 1, 3, 6 and 12. T2DM, Type 2 diabetes mellitus.
Figure 5
Figure 5
The immunologic tests and markers of systemic inflammation. A: The changes in the numbers of CD3+, CD4+ and CD8+ T lymphocytes between baseline and six months. B: Changes in the serum levels of IL-6, IL-10, IL-1β and TNF-α between baseline and six months. The results are shown as the mean ± S.D. *P <0.05 compared with pretreatment. The analysis included 22 patients before and six months after umbilical cord mesenchymal stem cell transplantation.

References

    1. Kolb H, Mandrup-Poulsen T. An immune origin of type 2 diabetes? Diabetologia. 2005;48:1038–1050.
    1. Tysseling-Mattiace VM, Sahni V, Niece KL, Birch D, Czeisler C, Fehlings MG, Stupp SI, Kessler JA. Self-assembling nanofibers inhibit glial scar formation and promote axon elongation after spinal cord injury. J Neurosci. 2008;28:3814–3823. doi: 10.1523/JNEUROSCI.0143-08.2008.
    1. Wright A, Burden AC, Paisey RB, Cull CA, Holman RR. Sulfonylurea inadequacy: efficacy of addition of insulin over 6 years in patients with type 2 diabetes in the U.K. Prospective Diabetes Study (UKPDS 57) Diabetes Care. 2002;25:330–336. doi: 10.2337/diacare.25.2.330.
    1. Bhansali A, Upreti V, Khandelwal N, Marwaha N, Gupta V, Sachdeva N, Sharma RR, Saluja K, Dutta P, Walia R, Minz R, Bhadada S, Das S, Ramakrishnan S. Efficacy of autologous bone marrow-derived stem cell transplantation in patients with type 2 diabetes mellitus. Stem Cells Dev. 2009;18:1407–1416. doi: 10.1089/scd.2009.0164.
    1. Jiang R, Han Z, Zhuo G, Qu X, Li X, Wang X, Shao Y, Yang S, Han ZC. Transplantation of placenta-derived mesenchymal stem cells in type 2 diabetes: a pilot study. Front Med. 2011;5:94–100. doi: 10.1007/s11684-011-0116-z.
    1. Li XY, Zheng ZH, Guo J, Zhang Y, Li H, Wang YW, Ren J, Wu ZB. Treatment of foot disease in patients with type 2 diabetes mellitus using human umbilical cord blood mesenchymal stem cells: response and correction of immunological anomalies. Curr Pharm Des. 2013;19:4893–4899. doi: 10.2174/13816128113199990326.
    1. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112:1821–1830. doi: 10.1172/JCI200319451.
    1. Donath MY, Boni-Schnetzler M, Ellingsgaard H, Ehses JA. Islet inflammation impairs the pancreatic beta-cell in type 2 diabetes. Physiology (Bethesda) 2009;24:325–331. doi: 10.1152/physiol.00032.2009.
    1. Guo J, Lin GS, Bao CY, Hu ZM, Hu MY. Anti-inflammation role for mesenchymal stem cells transplantation in myocardial infarction. Inflammation. 2007;30:97–104. doi: 10.1007/s10753-007-9025-3.
    1. Moodley Y, Atienza D, Manuelpillai U, Samuel CS, Tchongue J, Ilancheran S, Boyd R, Trounson A. Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Pathol. 2009;175:303–313. doi: 10.2353/ajpath.2009.080629.
    1. Sun L, Wang D, Liang J, Zhang H, Feng X, Wang H, Hua B, Liu B, Ye S, Hu X, Xu W, Zeng X, Hou Y, Gilkeson GS, Silver RM, Lu L, Shi S. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis Rheum. 2010;62:2467–2475. doi: 10.1002/art.27548.
    1. Le Blanc K, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, Uzunel M, Ringden O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004;363:1439–1441. doi: 10.1016/S0140-6736(04)16104-7.
    1. Zappia E, Casazza S, Pedemonte E, Benvenuto F, Bonanni I, Gerdoni E, Giunti D, Ceravolo A, Cazzanti F, Frassoni F, Mancardi G, Uccelli A. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy. Blood. 2005;106:1755–1761. doi: 10.1182/blood-2005-04-1496.
    1. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2006;29:S43–S48.
    1. University of Oxford: The Diabetes Trial Centre for Diabetes, Endocrinology and Metabolism. [ ]
    1. Wallace TM, Levy JC, Matthews DR. Use and abuse of HOMA modeling. Diabetes Care. 2004;27:1487–1495. doi: 10.2337/diacare.27.6.1487.
    1. Rohlfing CL, Wiedmeyer HM, Little RR, England JD, Tennill A, Goldstein DE. Defining the relationship between plasma glucose and HbA(1c): analysis of glucose profiles and HbA(1c) in the Diabetes Control and Complications Trial. Diabetes Care. 2002;25:275–278. doi: 10.2337/diacare.25.2.275.
    1. Spranger J, Kroke A, Mohlig M, Hoffmann K, Bergmann MM, Ristow M, Boeing H, Pfeiffer AF. Inflammatory cytokines and the risk to develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes. 2003;52:812–817. doi: 10.2337/diabetes.52.3.812.
    1. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest. 2005;115:1111–1119. doi: 10.1172/JCI200525102.
    1. Wei Y, Chen K, Whaley-Connell AT, Stump CS, Ibdah JA, Sowers JR. Skeletal muscle insulin resistance: role of inflammatory cytokines and reactive oxygen species. Am J Physiol Regul Integr Comp Physiol. 2008;294:R673–R680. doi: 10.1152/ajpregu.00561.2007.
    1. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am J Physiol Endocrinol Metab. 2001;280:E745–E751.
    1. Welsh N, Cnop M, Kharroubi I, Bugliani M, Lupi R, Marchetti P, Eizirik DL. Is there a role for locally produced interleukin-1 in the deleterious effects of high glucose or the type 2 diabetes milieu to human pancreatic islets? Diabetes. 2005;54:3238–3244. doi: 10.2337/diabetes.54.11.3238.
    1. Lee H, Bae JS, Jin HK. Human umbilical cord blood-derived mesenchymal stem cells improve neurological abnormalities of Niemann-Pick type C mouse by modulation of neuroinflammatory condition. J Vet Med Sci. 2010;72:709–717. doi: 10.1292/jvms.09-0495.
    1. Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, Mandrup-Poulsen T, Donath MY. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356:1517–1526. doi: 10.1056/NEJMoa065213.
    1. Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, Dorfman R, Wang Y, Zielenski J, Mastronardi F, Maezawa Y, Drucker DJ, Engleman E, Winer D, Dosch HM. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med. 2009;15:921–929. doi: 10.1038/nm.2001.
    1. Kintscher U, Hartge M, Hess K, Foryst-Ludwig A, Clemenz M, Wabitsch M, Fischer-Posovszky P, Barth TF, Dragun D, Skurk T, Hauner H, Bluher M, Unger T, Wolf AM, Knippschild U, Hombach V, Marx N. T-lymphocyte infiltration in visceral adipose tissue: a primary event in adipose tissue inflammation and the development of obesity-mediated insulin resistance. Arterioscler Thromb Vasc Biol. 2008;28:1304–1310. doi: 10.1161/ATVBAHA.108.165100.
    1. Voltarelli JC, Couri CE, Stracieri AB, Oliveira MC, Moraes DA, Pieroni F, Coutinho M, Malmegrim KC, Foss-Freitas MC, Simoes BP, Foss MC, Squiers E, Burt RK. Autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA. 2007;297:1568–1576. doi: 10.1001/jama.297.14.1568.
    1. Lama VN, Smith L, Badri L, Flint A, Andrei AC, Murray S, Wang Z, Liao H, Toews GB, Krebsbach PH, Peters-Golden M, Pinsky DJ, Martinez FJ, Thannickal VJ. Evidence for tissue-resident mesenchymal stem cells in human adult lung from studies of transplanted allografts. J Clin Invest. 2007;117:989–996. doi: 10.1172/JCI29713.
    1. Hu J, Yu X, Wang Z, Wang F, Wang L, Gao H, Chen Y, Zhao W, Jia Z, Yan S, Wang Y. Long term effects of the implantation of Wharton's jelly-derived mesenchymal stem cells from the umbilical cord for newly-onset type 1 diabetes mellitus. Endocr J. 2013;60:347–357. doi: 10.1507/endocrj.EJ12-0343.
    1. Jin JL, Liu Z, Lu ZJ, Guan DN, Wang C, Chen ZB, Zhang J, Zhang WY, Wu JY, Xu Y. Safety and efficacy of umbilical cord mesenchymal stem cell therapy in hereditary spinocerebellar ataxia. Curr Neurovasc Res. 2013;10:11–20. doi: 10.2174/156720213804805936.

Source: PubMed

3
Subscribe