Autologous pancreatic islet transplantation in human bone marrow

Paola Maffi, Gianpaolo Balzano, Maurilio Ponzoni, Rita Nano, Valeria Sordi, Raffaella Melzi, Alessia Mercalli, Marina Scavini, Antonio Esposito, Jacopo Peccatori, Elisa Cantarelli, Carlo Messina, Massimo Bernardi, Alessandro Del Maschio, Carlo Staudacher, Claudio Doglioni, Fabio Ciceri, Antonio Secchi, Lorenzo Piemonti, Paola Maffi, Gianpaolo Balzano, Maurilio Ponzoni, Rita Nano, Valeria Sordi, Raffaella Melzi, Alessia Mercalli, Marina Scavini, Antonio Esposito, Jacopo Peccatori, Elisa Cantarelli, Carlo Messina, Massimo Bernardi, Alessandro Del Maschio, Carlo Staudacher, Claudio Doglioni, Fabio Ciceri, Antonio Secchi, Lorenzo Piemonti

Abstract

The liver is the current site of choice for pancreatic islet transplantation, even though it is far from being ideal. We recently have shown in mice that the bone marrow (BM) may be a valid alternative to the liver, and here we report a pilot study to test feasibility and safety of BM as a site for islet transplantation in humans. Four patients who developed diabetes after total pancreatectomy were candidates for the autologous transplantation of pancreatic islet. Because the patients had contraindications for intraportal infusion, islets were infused in the BM. In all recipients, islets engrafted successfully as shown by measurable posttransplantation C-peptide levels and histopathological evidence of insulin-producing cells or molecular markers of endocrine tissue in BM biopsy samples analyzed during follow-up. Thus far, we have recorded no adverse events related to the infusion procedure or the presence of islets in the BM. Islet function was sustained for the maximum follow-up of 944 days. The encouraging results of this pilot study provide new perspectives in identifying alternative sites for islet infusion in patients with type 1 diabetes. Moreover, this is the first unequivocal example of successful engraftment of endocrine tissue in the BM in humans.

Figures

FIG. 1.
FIG. 1.
Metabolic control and graft function after IAT. Insulin requirement and fasting C-peptide after IAT. Data are shown for each individual patient. Left axis: Exogenous insulin requirement expressed as IU/kg/day. Right axis: Fasting C-peptide expressed as ng/mL. The gray area represents the adjuvant therapy treatment for primary cancer during follow-up (chemotherapy or radiotherapy or both). Dotted line represents median fasting C-peptide (interquartile range represented by the yellow area) of 13 consecutive patients with total pancreatectomy receiving intraportal autologous islet infusion (median, 1,939 IEQ/kg; 25–75th percentiles, 1,659–2,151 IEQ/kg; min-max, 628–3,154 IEQ/kg) at our center during the same period. □, insulin requirement; ●, C-peptide.
FIG. 2.
FIG. 2.
BM morphology at day 4 posttransplantation. Photomicrographs of BM tissue obtained from the postmortem examination of patient 1. A: Histological appearance (left panel, magnification 100×; right panel, inset of left panel, magnification 400×) of transplanted tissue. Hematoxylin and eosin staining. Str, stromal reaction; Is, pancreatic islet; Epit, epithelial pancreatic cells. B: Representative immunohistochemical stainings (magnification 200×) with anti-insulin, antiglucagon, anticytokeratin 8–18 (cytokeratin 8–18), and anti-CD34 antibodies.
FIG. 3.
FIG. 3.
BM morphology at 1 year posttransplantation. Photomicrographs of BM biopsy samples from patient 3. A: Histological appearance (left panel, magnification 200×; right panel, inset of left panel, magnification 400×) of transplanted tissue. Hematoxylin and eosin staining. B: Representative immunohistochemical stainings (magnification 200×) with anti-insulin, antiglucagon, antichromogranin A, antisomatostatin, antipancreatic polypeptide, and anti-CD34 antibodies.
FIG. 4.
FIG. 4.
BM biopsy molecular assay. Scatter plot comparing the normalized expression (2−ΔCt) of every gene with graft-bearing and contralateral BM. The central line indicates unchanged gene expression and the dashed line indicates the boundary (fold regulation cutoff set = 3). Expression changes greater than the selected boundary (filled circles) and expression changes smaller than the selected boundary (gray circles) are shown.
FIG. 5.
FIG. 5.
MRI and computed tomography scan of iliac BM. MRI T2-weighted images acquired 30 and 90 days and 1 year after islet transplantation in patient 3 (left). A small hypointense area (red circle) inside the normal hyperintense signal was evident at the site of the islet infusion at the level of the posterior-superior iliac spine. The gadolinium-enhanced MR perfusion study did not show areas of anomalous enhancement surrounding the site of the islets infusion (right). A computed tomography scan showed the presence of a small calcified spot at the site of islet infusion (lower right).

References

    1. Barton FB, Rickels MR, Alejandro R, et al. Improvement in outcomes of clinical islet transplantation: 1999-2010. Diabetes Care 2012;35:1436–1445
    1. Toyofuku A, Yasunami Y, Nabeyama K, et al. Natural killer T-cells participate in rejection of islet allografts in the liver of mice. Diabetes 2006;55:34–39
    1. Yasunami Y, Kojo S, Kitamura H, et al. Valpha14 NK T cell-triggered IFN-gamma production by Gr-1+CD11b+ cells mediates early graft loss of syngeneic transplanted islets. J Exp Med 2005;202:913–918
    1. Citro A, Cantarelli E, Maffi P, et al. CXCR1/2 inhibition enhances pancreatic islet survival after transplantation. J Clin Invest 2012;122:3647–3651
    1. Korsgren O, Lundgren T, Felldin M, et al. Optimising islet engraftment is critical for successful clinical islet transplantation. Diabetologia 2008;51:227–232
    1. Desai NM, Goss JA, Deng S, et al. Elevated portal vein drug levels of sirolimus and tacrolimus in islet transplant recipients: local immunosuppression or islet toxicity? Transplantation 2003;76:1623–1625
    1. Shapiro AM, Gallant HL, Hao EG, et al. The portal immunosuppressive storm: relevance to islet transplantation? Ther Drug Monit 2005;27:35–37
    1. Bhargava R, Senior PA, Ackerman TE, et al. Prevalence of hepatic steatosis after islet transplantation and its relation to graft function. Diabetes 2004;53:1311–1317
    1. Piemonti L, Everly MJ, Maffi P, et al. Alloantibody and autoantibody monitoring predicts islet transplantation outcome in human type 1 diabetes. Diabetes 2013;62:1656–1664
    1. Piemonti L, Guidotti LG, Battaglia M. Modulation of early inflammatory reactions to promote engraftment and function of transplanted pancreatic islets in autoimmune diabetes. Adv Exp Med Biol 2010;654:725–747
    1. Moberg L, Johansson H, Lukinius A, et al. Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet 2002;360:2039–2045
    1. Eich T, Eriksson O, Lundgren T, Nordic Network for Clinical Islet Transplantation Visualization of early engraftment in clinical islet transplantation by positron-emission tomography. N Engl J Med 2007;356:2754–2755
    1. Barshes NR, Wyllie S, Goss JA. Inflammation-mediated dysfunction and apoptosis in pancreatic islet transplantation: implications for intrahepatic grafts. J Leukoc Biol 2005;77:587–597
    1. Eriksson O, Eich T, Sundin A, et al. Positron emission tomography in clinical islet transplantation. Am J Transplant 2009;9:2816–2824
    1. Crowe LA, Ris F, Nielles-Vallespin S, et al. A novel method for quantitative monitoring of transplanted islets of langerhans by positive contrast magnetic resonance imaging. Am J Transplant 2011;11:1158–1168
    1. Bennet W, Sundberg B, Groth CG, et al. Incompatibility between human blood and isolated islets of Langerhans: a finding with implications for clinical intraportal islet transplantation? Diabetes 1999;48:1907–1914
    1. Johansson H, Lukinius A, Moberg L, et al. Tissue factor produced by the endocrine cells of the islets of Langerhans is associated with a negative outcome of clinical islet transplantation. Diabetes 2005;54:1755–1762
    1. Sakata N, Hayes P, Tan A, et al. MRI assessment of ischemic liver after intraportal islet transplantation. Transplantation 2009;87:825–830
    1. Yin D, Ding JW, Shen J, Ma L, Hara M, Chong AS. Liver ischemia contributes to early islet failure following intraportal transplantation: benefits of liver ischemic-preconditioning. Am J Transplant 2006;6:60–68
    1. Barshes NR, Lee TC, Goodpastor SE, et al. Transaminitis after pancreatic islet transplantation. J Am Coll Surg 2005;200:353–361
    1. Rafael E, Ryan EA, Paty BW, et al. Changes in liver enzymes after clinical islet transplantation. Transplantation 2003;76:1280–1284
    1. Casey JJ, Lakey JR, Ryan EA, et al. Portal venous pressure changes after sequential clinical islet transplantation. Transplantation 2002;74:913–915
    1. Morrison CP, Wemyss-Holden SA, Dennison AR, Maddern GJ. Islet yield remains a problem in islet autotransplantation. Arch Surg 2002;137:80–83
    1. Cantarelli E, Piemonti L. Alternative transplantation sites for pancreatic islet grafts. Curr Diab Rep 2011;11:364–374
    1. Ciceri F, Piemonti L. Bone marrow and pancreatic islets: an old story with new perspectives. Cell Transplant 2010;19:1511–1522
    1. Cantarelli E, Melzi R, Mercalli A, et al. Bone marrow as an alternative site for islet transplantation. Blood 2009;114:4566–4574
    1. Melzi R, Mercalli A, Sordi V, et al. Role of CCL2/MCP-1 in islet transplantation. Cell Transplant 2010;19:1031–1046
    1. Frassoni F, Gualandi F, Podestà M, et al. Direct intrabone transplant of unrelated cord-blood cells in acute leukaemia: a phase I/II study. Lancet Oncol 2008;9:831–839
    1. Caumo A, Maffi P, Nano R, et al. Comparative evaluation of simple indices of graft function after islet transplantation. Transplantation 2011;92:815–821
    1. Mercalli A, Sordi V, Ponzoni M, et al. Rapamycin induces a caspase-independent cell death in human monocytes. Am J Transplant 2006;6:1331–1341
    1. Bassi C, Dervenis C, Butturini G, et al. International Study Group on Pancreatic Fistula Definition Postoperative pancreatic fistula: an international study group (ISGPF) definition. Surgery 2005;138:8–13
    1. Toso C, Isse K, Demetris AJ, et al. Histologic graft assessment after clinical islet transplantation. Transplantation 2009;88:1286–1293
    1. Servitja JM, Ferrer J. Transcriptional networks controlling pancreatic development and beta cell function. Diabetologia 2004;47:597–613
    1. Kemp CB, Knight MJ, Scharp DW, Ballinger WF, Lacy PE. Effect of transplantation site on the results of pancreatic islet isografts in diabetic rats. Diabetologia 1973;9:486–491
    1. Scharp DW, Lacy PE, Santiago JV, et al. Insulin independence after islet transplantation into type I diabetic patient. Diabetes 1990;39:515–518
    1. Rafael E, Tibell A, Rydén M, et al. Intramuscular autotransplantation of pancreatic islets in a 7-year-old child: a 2-year follow-up. Am J Transplant 2008;8:458–462
    1. Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J, Kaupper T, Roncarolo MG. Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients. J Immunol 2006;177:8338–8347
    1. Tan J, Wu W, Xu X, et al. Induction therapy with autologous mesenchymal stem cells in living-related kidney transplants: a randomized controlled trial. JAMA 2012;307:1169–1177
    1. Piatti PM, Pontiroli AE, Caumo A, et al. Hyperinsulinemia decreases second-phase but not first-phase arginine-induced insulin release in humans. Diabetes 1994;43:1157–1163

Source: PubMed

3
Suscribir