Limitations of IL-2 and Rapamycin in Immunotherapy of Type 1 Diabetes

Administration of low-dose interleukin-2 (IL-2) alone or combined with rapamycin (RAPA) prevents hyperglycemia in NOD mice. Also, low-dose IL-2 cures recent-onset type 1 diabetes (T1D) in NOD mice, partially by boosting pancreatic regulatory T cells (Treg cells). These approaches are currently being evaluated in humans. Our objective was to study the effect of higher IL-2 doses (250,000–500,000 IU daily) as well as low-dose IL-2 (25,000 IU daily) and RAPA (1 mg/kg daily) (RAPA/IL-2) combination. We show that, despite further boosting of Treg cells, high doses of IL-2 rapidly precipitated T1D in prediabetic female and male mice and increased myeloid cells in the pancreas. Also, we observed that RAPA counteracted IL-2 effects on Treg cells, failed to control IL-2–boosted NK cells, and broke IL-2–induced tolerance in a reversible way. Notably, the RAPA/IL-2 combination failure to cure T1D was associated with an unexpected deleterious effect on glucose homeostasis at multiple levels, including β-cell division, glucose tolerance, and liver glucose metabolism. Our data help to understand the therapeutic limitations of IL-2 alone or RAPA/IL-2 combination and could lead to the design of improved therapies for T1D.

[1]  J. Bluestone,et al.  Rapamycin/IL-2 Combination Therapy in Patients With Type 1 Diabetes Augments Tregs yet Transiently Impairs β-Cell Function , 2012, Diabetes.

[2]  J. Bluestone,et al.  NK Cells Are Not Required for Spontaneous Autoimmune Diabetes in NOD Mice , 2012, PloS one.

[3]  Dudley Lamming,et al.  Rapamycin-Induced Insulin Resistance Is Mediated by mTORC2 Loss and Uncoupled from Longevity , 2012, Science.

[4]  R. Weissleder,et al.  Early window of diabetes determinism in NOD mice, dependent on the complement receptor CRIg, identified by noninvasive imaging , 2012, Nature Immunology.

[5]  L. Wen,et al.  The Role of Gr1+ Cells after Anti-CD20 Treatment in Type 1 Diabetes in Nonobese Diabetic Mice , 2012, The Journal of Immunology.

[6]  J. Ritz,et al.  Interleukin-2 and regulatory T cells in graft-versus-host disease. , 2011, The New England journal of medicine.

[7]  F. Carrat,et al.  Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. , 2011, The New England journal of medicine.

[8]  Y. Dor,et al.  Recognition and Killing of Human and Murine Pancreatic β Cells by the NK Receptor NKp46 , 2011, The Journal of Immunology.

[9]  R. Negrin,et al.  Rapamycin and IL-2 reduce lethal acute graft-versus-host disease associated with increased expansion of donor type CD4+CD25+Foxp3+ regulatory T cells. , 2011, Blood.

[10]  J. Bach Anti-CD3 antibodies for type 1 diabetes: beyond expectations , 2011, The Lancet.

[11]  K. Herold,et al.  Immunomodulatory therapy to preserve pancreatic β-cell function in type 1 diabetes , 2011, Nature Reviews Drug Discovery.

[12]  C. Benoist,et al.  A cluster of coregulated genes determines TGF-β–induced regulatory T-cell (Treg) dysfunction in NOD mice , 2011, Proceedings of the National Academy of Sciences.

[13]  E. Bonifacio,et al.  Beta cell function during rapamycin monotherapy in long-term type 1 diabetes , 2011, Diabetologia.

[14]  D. Klatzmann,et al.  Pathogenic T cells have a paradoxical protective effect in murine autoimmune diabetes by boosting Tregs. , 2010, The Journal of clinical investigation.

[15]  F. Vély,et al.  Natural killer cells in human autoimmune diseases , 2010, Immunology.

[16]  C. Divino,et al.  Myeloid-Derived Suppressor Cells Prevent Type 1 Diabetes in Murine Models , 2010, The Journal of Immunology.

[17]  J. Bluestone,et al.  IL-2 reverses established type 1 diabetes in NOD mice by a local effect on pancreatic regulatory T cells , 2010, The Journal of experimental medicine.

[18]  G. Pantaleo,et al.  Improved IL-2 immunotherapy by selective stimulation of IL-2 receptors on lymphocytes and endothelial cells , 2010, Proceedings of the National Academy of Sciences.

[19]  Jeffrey A. Bluestone,et al.  Genetics, pathogenesis and clinical interventions in type 1 diabetes , 2010, Nature.

[20]  Y. Deshaies,et al.  Chronic Rapamycin Treatment Causes Glucose Intolerance and Hyperlipidemia by Upregulating Hepatic Gluconeogenesis and Impairing Lipid Deposition in Adipose Tissue , 2010, Diabetes.

[21]  Y. Dor,et al.  The activating receptor NKp46 is essential for the development of type 1 diabetes , 2010, Nature Immunology.

[22]  C. Mathieu,et al.  Four-year metabolic outcome of a randomised controlled CD3-antibody trial in recent-onset type 1 diabetic patients depends on their age and baseline residual beta cell mass , 2010, Diabetologia.

[23]  C. Benoist,et al.  How punctual ablation of regulatory T cells unleashes an autoimmune lesion within the pancreatic islets. , 2009, Immunity.

[24]  P. Worley,et al.  The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. , 2009, Immunity.

[25]  A. Thomson,et al.  Immunoregulatory functions of mTOR inhibition , 2009, Nature Reviews Immunology.

[26]  M. Atkinson,et al.  Rapamycin Prevents and Breaks the Anti-CD3–Induced Tolerance in NOD Mice , 2009, Diabetes.

[27]  E. Bonifacio,et al.  Rapamycin Monotherapy in Patients With Type 1 Diabetes Modifies CD4+CD25+FOXP3+ Regulatory T-Cells , 2008, Diabetes.

[28]  A. Webster,et al.  Sirolimus is associated with new-onset diabetes in kidney transplant recipients. , 2008, Journal of the American Society of Nephrology : JASN.

[29]  J. Bluestone,et al.  Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. , 2008, Immunity.

[30]  C. Benoist,et al.  The AKT–mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells , 2008, The Journal of experimental medicine.

[31]  R. Roberts Faculty Opinions recommendation of mTOR controls mitochondrial oxidative function through a YY1-PGC-1alpha transcriptional complex. , 2007 .

[32]  V. Mootha,et al.  mTOR controls mitochondrial oxidative function through a YY1–PGC-1α transcriptional complex , 2007, Nature.

[33]  D. Klatzmann,et al.  Regulatory and Effector T Cell Activation Levels Are Prime Determinants of In Vivo Immune Regulation1 , 2006, The Journal of Immunology.

[34]  R. Liblau,et al.  Pertussis Toxin Reduces the Number of Splenic Foxp3+ Regulatory T Cells1 , 2006, The Journal of Immunology.

[35]  E. Bonifacio,et al.  Induction of Tolerance in Type 1 Diabetes via Both CD4+CD25+ T Regulatory Cells and T Regulatory Type 1 Cells , 2006, Diabetes.

[36]  T. Waldmann,et al.  Regulatory CD56(bright) natural killer cells mediate immunomodulatory effects of IL-2Ralpha-targeted therapy (daclizumab) in multiple sclerosis. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[37]  M. Battaglia,et al.  Rapamycin selectively expands CD4+CD25+FoxP3+ regulatory T cells. , 2005, Blood.

[38]  R. Nussenblatt,et al.  Cutting Edge: In Vivo Blockade of Human IL-2 Receptor Induces Expansion of CD56bright Regulatory NK Cells in Patients with Active Uveitis , 2005, The Journal of Immunology.

[39]  D. Finegood,et al.  Prediction of spontaneous autoimmune diabetes in NOD mice by quantification of autoreactive T cells in peripheral blood. , 2003, The Journal of clinical investigation.

[40]  A. Shapiro,et al.  Combination therapy with sirolimus and interleukin-2 prevents spontaneous and recurrent autoimmune diabetes in NOD mice. , 2002, Diabetes.

[41]  A. M. Shapiro,et al.  Combination therapy with low dose sirolimus and tacrolimus is synergistic in preventing spontaneous and recurrent autoimmune diabetes in non-obese diabetic mice , 2002, Diabetologia.

[42]  M. Atkinson,et al.  The NOD mouse model of type 1 diabetes: As good as it gets? , 1999, Nature Medicine.

[43]  K. Polonsky,et al.  Increased beta-cell proliferation and reduced mass before diabetes onset in the nonobese diabetic mouse. , 1999, Diabetes.

[44]  H. Drexhage,et al.  Immunohistochemical Characterization of Monocytes-Macrophages and Dendritic Cells Involved in the Initiation of the Insulitis and β-Cell Destruction in NOD Mice , 1994, Diabetes.

[45]  L. Chatenoud,et al.  Anti-CD3 antibody induces long-term remission of overt autoimmunity in nonobese diabetic mice. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[46]  J. Sredy,et al.  Rapamycin prevents the onset of insulin‐dependent diabetes mellitus (IDDM) in NOD mice , 1992, Clinical and experimental immunology.

[47]  G. Crabtree,et al.  Rapamycin selectively inhibits interleukin-2 activation of p70 S6 kinase , 1992, Nature.

[48]  M. V. von Herrath,et al.  Type 1 diabetes: etiology, immunology, and therapeutic strategies. , 2011, Physiological reviews.

[49]  R. Abraham,et al.  Immunopharmacology of rapamycin. , 1996, Annual review of immunology.

[50]  D. Monos,et al.  Insulin dependent diabetes mellitus as an autoimmune disease. , 1988, In vivo.