Horm Metab Res 2015; 47(10): 797-803
DOI: 10.1055/s-0035-1555956
Review

Modeling Graves’ Orbitopathy in Experimental Graves’ Disease

J. P. Banga
1   Faculty of Life Sciences & Medicine, King’s College London, The Rayne Institute, London, UK
2   Department of Ophthalmology, University of Duisburg-Essen, Essen, Germany
,
S. Moshkelgosha
1   Faculty of Life Sciences & Medicine, King’s College London, The Rayne Institute, London, UK
2   Department of Ophthalmology, University of Duisburg-Essen, Essen, Germany
,
U. Berchner-Pfannschmidt
2   Department of Ophthalmology, University of Duisburg-Essen, Essen, Germany
,
A. Eckstein
2   Department of Ophthalmology, University of Duisburg-Essen, Essen, Germany
› Author Affiliations

Abstract

Graves’ orbitopathy (GO), also known as thyroid eye disease is an inflammatory disease of the orbital tissue of the eye that arises as a consequence of autoimmune thyroid disease. The central feature of the disease is the production of antibodies to the thyrotropin hormone receptor (TSHR) that modulate the function of the receptor leading to autoimmune hyperthyroidism and GO. Over the years, all viable preclinical models of Graves’ disease have been incomplete and singularly failed to progress in the treatment of orbital complications. A new mouse model of GO based upon immunogenic presentation of human TSHR A-subunit plasmid by close field electroporation is shown to lead to induction of prolonged functional antibodies to TSHR resulting in chronic disease with subsequent progression to GO. The stable preclinical GO model exhibited pathologies reminiscent of human disease characterized by orbital remodeling by inflammation and adipogenesis. Inflammatory lesions characterized by CD3+ T cells and macrophages were localized in the orbital muscle tissue. This was accompanied by extensive adipogenesis of orbital fat in some immune animals. Surprisingly, other signs of orbital involvement were reminiscent of eyelid inflammation involving chemosis, with dilated and congested orbital blood vessels. More recently, the model is replicated in the author’s independent laboratories. The pre-clinical model will provide the basis to study the pathogenic and regulatory roles of immune T and B cells and their subpopulations to understand the initiation, pathophysiology, and progression of GO.



Publication History

Received: 20 April 2015

Accepted: 08 July 2015

Article published online:
19 August 2015

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Bartelena L. Diagnosis and management of Graves’ disease: a global overview. Nature Rev Endocrinol 2013; 9: 723-734
  • 2 Weetman AP. Medical progress: Graves’ disease. New Engl J Med 2000; 343: 1236-1238
  • 3 Bahn RS. Graves’ ophthalmopathy. New Engl J Med 2010; 362: 726-738
  • 4 Shan SJC, Douglas RS. The pathophysiology of thyroid eye disease. J neuroophthalmol 2014; 34: 177-185
  • 5 Regensburg NI, Wiersinga WM, Berendschot TT, Potgieser P, Mourits MP. Do subtypes of graves’ orbitopathy exist?. Ophthalmol 2011; 118: 191-196
  • 6 Leo M, Menconi F, Rocchi R, Latrofa F, Sisti E, Profilo MA, Mazzi B, Albano E, Nardi M, Vitti P, Marcocci C, Marino M. Role of underlying thyroid disease on the phenotype of Graves’ orbitopathy in a tertiary referral center. Thyroid 2015; 25: 347-351
  • 7 Heufelder AE, Smith TJ, Gorman CA, Bahn RS. Increased induction of HLA-DR in interferon-gamma in cultured fibroblasts derived from patients with Graves’ ophthalmopathy and pretibial dermopathy. J Clin Endocrinol Metab 1991; 73: 307-313
  • 8 Sorisky A, Pardasani D, Gagnon A, Smith TJ. Evidence of adipocyte differentiation in human orbital fibroblasts in primary culture. J Clin Endocrinol Metab 1996; 81: 3428-33431
  • 9 Bahn RS, Dutton CM, Natt N, Joba W, Spitzweg C, Heufelder AE. Thyrotropin receptor expression in Graves’ orbital adipose/connective tissues: potential autoantigen in Graves’ ophthalmopathy. J Clin Endocrinol Metab 1998; 83: 998-1002
  • 10 Wall JR. Thyroid function. Pathogenesis of Graves’ ophthalmopathy – a role for TSH-R?. Nat Revs Endocrinol 2014; 10: 256-268
  • 11 Smith TJ. The putative role of fibroblasts in the pathogenesis of Graves’ disease: Evidence for the involvement of the insulin-like growth factor-1 receptor in fibroblast activation. Autoimmunity 2003; 36: 409-415
  • 12 Smith TJ. Potential role for bone marrow-derived fibrocytes in the orbital fibroblast heterogeneity associated with thyroid-associated ophthalmopathy. Clin Exp Immunol 2010; 162: 24-31
  • 13 Kumar S, Iyer S, Bauer H, Coenen M, Bahn RS. A stimulatory thyrotropin antibody enhances hyaluronic acid synthesis in Graves’ orbital fibroblasts: inhibition by an IGF-1 receptor blocking antibody. J Clin Endocrinol Metab 2012; 97: 1681-1687
  • 14 van Zeijl CJ, van Koppen CJ, Surovtseva OV, de Gooyer ME, Plate R, Conti P, Karstens WJ, Timmers M, Saeed P, Wiersinga WM, Miltenburg AM, Fliers E, Boelen A. Complete inhibition of rhTSH-, Graves’ disease IgG-, and M22-induced cAMP production in differentiated orbital fibroblasts by a low-molecular-weight TSHR antagonist. J Clin Endocrinol Metab 2012; 97: E781-E785
  • 15 Krieger CC, Neumann S, Place RF, Marcus-Samuels B, Gershengorn MC. Bidirectional TSH and IGF-1receptor cross talk mediates stimulation of hyaluronan secretion by Graves’ disease immunoglobulins. J Clin Endocrinol Metab 2015; 100: 1071-1077
  • 16 Wiersinga W. Autoimmunity in Graves’ ophthalmopathy: the result of an unfortunate marriage between TSH receptors and IGF-1 receptors?. J Clin Endocrinol Metab 2012; 96: 2386-2394
  • 17 Dağdelen S, Kong Y-cM, Banga JP. Towards Better Models of Hyperthyroid Graves’ Disease. Endocrinol and Metab Clinics of North America 2009; 38: 343-354
  • 18 Wiesweg B, Johnson KT, Eckstein AK, Berchner-Pfannschmidt U. Current Insights into animal models of Graves’ disease and orbitopathy. Horm Metab Res 2013; 45: 549-555
  • 19 McLachlan SM, Alpi K, Rapoport B. Review and Hypothesis: Does Graves’ disease develop in non-human apes. Thyroid 2011; 21: 1359-1366
  • 20 Kriss JP, Pleshakov V, Chien JR. Isolation and identification of the long-acting thyroid stimulator and its relation to hyperthyroidism and circumscribed pretibial myxedema. J Clin Endocrinol 1964; 24: 1005-1028
  • 21 Rapoport B, Chazenbalk GD, Jaume JC, McLachlan SM, Rapoport B. The thyrotropin (TSHR receptor: interaction with TSH and autoantibodies. Endocrine Revs 1998; 18: 673-716
  • 22 Kajita Y, Rickards CR, Buckland PR, Howells RD, Rees Smith B. Analysis of thyrotropin receptors by photoaffinity labelling. Orientation of receptor subunits in the cell membrane. Biochem J 1985; 227: 413-420
  • 23 Loosfelt H, Pichon C, Jolivet A, Misrahi M, Caillou B, Jamous M, Vannier B, Milgrom E. Two-subunit structure of the human thyrotropin receptor. Proc Natl Acad Sci U S A 1992; 89: 3765-3769
  • 24 Tanaka K, Chazenbalk GD, McLachln SM, Rapoport B. Thyrotropin receptor cleavage at site 1 involves two discontinuous segments at each end of the unique 50-amino acid insertion. J Biol Chem 1999; 274: 2093-2096
  • 25 Kleinau G, Newmann S, Gruters A, Krude H, Biebermann H. Novel insights in thyroid-stimulating hormone receptor signal transduction. 2013; 34: 691-724
  • 26 Couet J, Sar S, Jolivet A, Hai MT, Milgrom E, Misrahi M. Shedding of human thyrotropin receptor ectodomain. Involvement of a matrix metalloprotease. J Biol Chem 1996; 271: 4545-4552
  • 27 Chen CR, Pichurin P, Nagayama Y, Latrofa F, Rapoport B, McLachlan SM. The thyrotropin receptor autoantigen in Graves disease is the culprit as well as the victim. J Clin Invest 2003; 111: 1897-1904
  • 28 Mizutori Y, Chen CR, Latrofa F, McLachlan SM, Rapoport B. Evidence that shed TSH receptor A-subunits drive affinity maturation of autoantibodies causing Graves’ disease. J Clin Endocrinol Metab 2009; 94: 927-935
  • 29 Rapoport B, Aliesky HA, Chen CR, McLachlan SM. Evidence that TSH receptor A-subunit multimers, not monomers, drive antibody affinity maturation in Graves’ disease. J Clin Endocrinol Metab 2015; 100: E871-E875
  • 30 Sanders J, Evans M, Premawardhana LDKE, Depraetere H, Jeffreys J, Richards T, Furmaniak J, Rees Smith B. Human monoclonal thyroid stimulating autoantibody. Lancet 2003; 362: 126-128
  • 31 Sanders J, Evans M, Betterle C, Sanders P, Bhardwaja A, Young S, Roberts E, Wilmot J, Richards T, Kiddie A, Small K, Platt H, Summerhayes S, Harris R, Reeve M, Coco G, Zanchetta R, Chen S, Furmaniak J, Rees Smith B. A human monoclonal autoantibody to the thyrotropin receptor with thyroid-stimulating blocking activity. Thyroid 2008; 18: 735-746
  • 32 Evans M, Sanders J, Tagami T, Sanders P, Young S, Roberts E, Wilmot J, Hu X, Kabelis K, Clark J, Holl S, Richards T, Collyer A, Furmaniak J, Smith BR. Monoclonal autoantibodies to the TSH receptor, one with stimulating activity and one with blocking activity, obtained from the same blood sample. Clin Endocrinol 2010; 73: 404-412
  • 33 Sanders J, Chirgadze DY, Sanders P, Baker S, Sullivan A, Bhardwaja A, Bolton J, Reeve M, Nakatake N, Evans M, Richards T, Powell M, Nunez Miguel R, Blundell TL, Furmaniak J, Rees Smith B. Crystal structure of the TSH receptor in complex with a thyroid-stimulating autoantibody. Thyroid 2007; 17: 395-410
  • 34 Sanders P, Young S, Sanders J, Kabelis K, Baker S, Sullivan A, Evans M, Clark J, Wilmot J, Hu X, Roberts E, Powell M, Nunez Miguel R, Furmaniak J, Rees Smith B. Crystal structure of the TSH receptor (TSHR) bound to a blocking-type TSHR autoantibody. J Mol Endocrinol 2011; 46: 81-99
  • 35 Nunez Miguel R, Sanders J, Sanders P, Young S, Clark J, Kabelis K, Wilmot J, Evans M, Roberts E, Hu X, Furmaniak J, Rees Smith B. Similarities and differences in interactions of thyroid stimulating and blocking autoantibodies with the TSH receptor. J Mol Endocrinol 2012; 49: 137-151
  • 36 Chen CR, Hubbard PA, Salazar LM, McLachlan SM, Rapoport B. Crystal structure of a TSH receptor monoclonal antibody: Insight into Graves’ disease pathogenesis. Mol Endocrinol 2015; 29: 99-107
  • 37 Neumann S, Kleinau G, Costanzi S, Moore S, Jiang JK, Raaka BM, Thomas CJ, Krause G, Gershengorn MC. A low-molecular-weight antagonist for the human thyrotropin receptor with therapeutic potential for hyperthyroidism. Endocrinology 2008; 149: 5945-5950
  • 38 Neumann S, Huang W, Eliseeva E, Titus S, Thomas CJ, Gershengorn MC. A small molecule inverse agonist for the human thyroid-stimulating hormone receptor. Endocrinology 2010; 151: 3454-3459
  • 39 van Koppen CJ, de Gooyer ME, Karstens WJ, Plate R, Conti PG, van Achterberg TA, van Amstel MG, Brands JH, Wat J, Berg RJ, Lane JR, Miltenburg AM, Timmers CM. Mechanism of action of a nanomolar potent, allosteric antagonist of the thyroid-stimulating hormone receptor. Br J Pharmacol 2012; 165: 2314-2324
  • 40 Turcu AF, Kumar S, Neumann S, Coenen M, Iyer S, Chiriboga P, Gershengorn MC, Bahn RS. A small molecule antagonist inhibits thyrotropin receptor antibody-induced orbital fibroblast functions involved in the pathogenesis of Graves ophthalmopathy. J Clin Endocrinol Metab 2013; 98: 2153-2159
  • 41 Latif R, Ali MR, Ma R, Morshed SA, Ohlmeyer M, Felsenfeld DP, Lau Z, Mezel M, Davies TF. New small molecule agonists to the thyrotropin receptor. Thyroid 2015; 25: 51-62
  • 42 Neumann S, Nir EA, Eliseeva E, Huang W, Marugan J, Xiao J, Dulcey AE, Gershengorn MC. A selective TSH receptor antagonist inhibits stimulation of thyroid function in female mice. Endocrinol 2014; 155: 310-314
  • 43 Banga JP, Nielsen CH, Gilbert JA, El Fassi D, Hegedus L. Application of new therapies in Graves' disease and thyroid-associated ophthalmopathy: animal models and translation to human clinical trials. Thyroid 2008; 18: 973-981
  • 44 Emerson CH. When will thyrotropin receptor antagonists and inverse thyrotropin receptor agonists become available for clinical use. Thyroid 2011; 21: 817-819
  • 45 Gershengorn MC, Neumann S. Update in TSH receptor agonists and antagonists. J Clin Endocrinol Metab 2012; 97: 4287-4292
  • 46 McLachlan SM, Nagayama Y, Rapoport B. Insight into Graves’ hyperthyroidism from animal models. Endocr Rev 2005; 26: 800-832
  • 47 Banga JP. The long and winding road for an experimental model of hyperthyroid Graves’ disease. In: Wiersinga WM, Drexhage HA, Weetman AP (eds.). The Thyroid and Autoimmunity. Stuttgart: Thieme Verlag; 2007: 118–125
  • 48 Carayanniotis G, Huang GC, Nicholson LB, Scott T, Allain P, McGregor AM, Banga JP. Unaltered thyroid function in mice responding to a highly immunogenic thyrotropin receptor: implications for the establishment of a mouse model for Graves’ disease. Clin Exp Immunol 1995; 99: 294-302
  • 49 Shimojo N, Kohno Y, Yamaguchi K, Kikuoka S, Hoshioka A, Niimi H, Hirai A, Tamura Y, Saito Y, Kohn LD, Tahara K. Induction of Graves’-like disease in mice by immunization with fibroblasts transfected with the thyrotropin receptor and a class II molecule. Proc Natl Acad Sci U S A 1996; 93: 11074-11079
  • 50 Kaithamana S, Fan J, Osuga Y, Liang SG, Prabhakar BS. Induction of experimental autoimmune Graves’ disease in BALB/c mice. J Immunol 1999; 163: 5157-5164
  • 51 Kita M, Ahmad L, Marians RC, Vlase H, Unger P, Graves PN, Davies TF. Regulation and transfer of a murine model of thyrotropin receptor antibody mediated Graves’ disease. Endocrinol 1999; 140: 1392-1398
  • 52 Ando T, Imaizumi M, Graves P. et al. Induction of thyroid stimulating hormone receptor autoimmunity in hamsters. Endocrinol 2003; 144: 671-860
  • 53 Land KJ, Moll JS, Kaplan MH. et al. Signal transducer and activator of transcription (Stat)-6-dependent, but not Stat-4-dependent, immunity is required for the development of autoimmunity in Graves’ hyperthyroidism. Endocrinol 2004; 145: 3724-3730
  • 54 Costagliola S, Rodien P, Many MC. et al. Genetic immunization against the human thyrotropin receptor causes thyroiditis and allows production of monoclonal antibodies recognizing the native receptor. J Immunol 1998; 160: 1458-1465
  • 55 Pichurin P, Yan XM, Farilla L, Guo J, Chazenbalk GD, Rapoport B, McLachlan SM. Naked TSH receptor DNA vaccination: A TH1 T cell response in which interferon-gamma production, rather than antibody, dominates the immune response in mice. Endocrinol 2001; 142: 3530-3536
  • 56 Rao PV, Watson PF, Weetman AP, Carayanniotis G, Banga JP. Contrasting activities of thyrotropin receptor antibodies in experimental models of Graves' disease induced by injection of transfected fibroblasts or deoxyribonucleic acid vaccination. Endocrinol 2003; 144: 260-266
  • 57 Nagayama Y, Kita-Furuyama M, Ando T, Nakao K, Mizuguchi H, Hayakawa T, Eguchi K, Niwa M. A novel murine model of Graves' hyperthyroidism with intramuscular injection of adenovirus expressing the thyrotropin receptor. J Immunol 2002; 168: 2789-2784
  • 58 Chen CR, Pichurin P, Nagayama Y, Latrofa F, Rapoport B, McLachlan SM. The thyrotropin receptor autoantigen in Graves disease is the culprit as well as the victim. J Clin Invest 2003; 111: 1897-1904
  • 59 Mizutori Y, Saitoh O, Eguchi K, Nagayama Y. Adenovirus encoding the thyrotropin receptor A-subunit improves the efficacy of dendritic cell-induced Graves’ hyperthyroidism in mice. J Autoimmun 2006; 26: 32-36
  • 60 Gilbert JA, Gianoukakis AG, Salehi S, Moorhead J, Rao PV, Khan MZ, McGregor AM, Smith TJ, Banga JP. Monoclonal pathogenic antibodies to the thyroid-stimulating hormone receptor in Graves’ disease with potent thyroid-stimulating activity but differential blocking activity activate multiple signaling pathways. J Immunol 2006; 176: 5084-5092
  • 61 Johnson KT, Wiesweg B, Schott M, Ehlers M, Muller M, Minich WB, Nagayama Y, Gulbins E, Eckstein AK, Berchner-Pfannschmidt U. Examination of orbital tissues in murine models of Graves’ disease reveals expression of UCP-1 and the TSHR in retrobulbar adipose tissues. Horm Metab Res 2013; 45: 401-407
  • 62 Wu L, Xun L, Yang J, Xu L, Tian Z, Gao S, Zhang Y, Hou P, Shi B. Induction of murine neonatal tolerance against Graves’ disease using recombinant adenovirus expressing the TSH receptor A-subunit. Endocrinol 2011; 152: 1165-1171
  • 63 Ye F, Hou P, Wu X, Ma X, Gao L, Wu L, Xu L, Shi B. The significance of immune-related molecule expression profiles in an animal model of Graves’ disease. Autoimmun 2012; 45: 143-152
  • 64 McLachlan SM, Aliesky HA, Chen CR, Rapoport B. Role of self-tolerance and chronic stimulation in the long-term persistence of adenovirus-induced thyrotropin receptor antibodies in wild-type and transgenic mice. Thyroid 2012; 22: 931-937
  • 65 Holthoff HP, Goebel S, Li Z, Fasbender J, Reiman A, Zeibig S, Lohse MJ, Munch G, Ungerer M. Prolonged TSH receptor A-subunit immunization of female mice leads to a long term model of Graves’ disease and cardiac hypertrophy. Endocrinol 2015; 156: 1577-1589
  • 66 Veron P, Leborgne C, Monteilhet V, Boutin S, Martin S, Moullier P, Masurier C. Humoral and cellular capsid-specific immune responses to adeno-associated virus type 1 in randomized healthy donors. J Immunol 2012; 188: 6418-6424
  • 67 Many M-C, Costagliola S, Detrait M, Denef J-F, Vassart G, Ludgate M. Development of an animal model of autoimmune thyroid eye disease. J Immunol 1999; 162: 4966-4974
  • 68 Costagliola S, Many MC, Denef JF, Pohlenz J, Refetoff S, Vassart G. Genetic immunization of outbred mice with thyrotropin receptor cDNA provides a model of Graves’ disease. J Clin Invest 2000; 105: 803-811
  • 69 Baker G, Mazziotti G, von Ruhland C, Ludgate M. Reevaluating thyrotropin receptor-induced mouse models of Graves’ disease and ophthalmopathy. Endocrinol 2005; 146: 835-844
  • 70 Yamada M, Li AW, West KA, Chang CH, Wall JR. Experimental model for ophthalmopathy in BALB/c and outbred (CD-1) mice genetically immunized with G2s and the thyrotropin receptor. Autoimmun 2002; 35: 403-313
  • 71 Bahn RS. At long last, an animal model of Graves’ orbitopathy. Endocrinol 2013; 154: 2989-2991
  • 72 Kaneda T, Honda A, Hakozaki A, Fuse T, Muto A, Yoshida T. An improved Graves’ disease model established by using in vivo electroporation exhibited long-term immunity to hyperthyroidism in BALB/c mice. Endocrinol 2007; 148: 2335-2344
  • 73 Widera G, Austin M, Rabussay D, Goldbeck C, Barnett SW, Chen M, Leung L, Otten GR, Thudium K, Selby MJ, Ulmer JB. Increased DNA vaccine delivery and immunogenicity by electroporation in vivo . J Immunol 2000; 164: 4635-4640
  • 74 Cemazar M, Golzio M, Sersa G, Rols MP, Teissie J. Electrically-assisted nucleic acids delivery to tissues in vivo: Where do we stand?. Curr Pharm Design 2006; 12: 3817-3825
  • 75 Zhao S-X, Tsui S, Cheung A, Douglas RS, Smith TJ, Banga JP. Orbital fibrosis in a mouse model of Graves’ disease induced by genetic immunization of thyrotropin receptor cDNA. J Endocrinol 2011; 210: 369-377
  • 76 Moshkelgosha S, So PW, Deasy N, Diaz-Cano S, Banga JP. Cutting Edge: Retrobulbar Inflammation, Adipogenesis, and Acute Orbital Congestion in a Preclinical Female Mouse Model of Graves’ Orbitopathy Induced by Thyrotropin Receptor Plasmid-in vivo Electroporation. Endocrinol 2013; 154: 3008-3015
  • 77 Tramontano D, Cushing GW, Moses AC, Ingbar SH. Insulin-like growth factor-I stimulates the growth of rat thyroid cells in culture and synergizes the stimulation of DNA synthesis induced by TSH and Graves’ IgG. Endocrinol 1986; 119: 940-942
  • 78 Kohn LD, Alvarez F, Marcocci C, Kohn AD, Corda D, Hoffman WE, Tombaccini D, Valente WA, De Luca M, Santisteban P. Monoclonal antibody studies defining the origin and properties of autoantibodies in Graves’ disease. Ann N Y Acad Sci 1986; 475: 157-173
  • 79 Weightman DR, Perros P, Sherif IH, Kendall-Taylor P. Autoantibodies to IGF-1 binding sites in thyroid associated ophthalmopathy. Autoimmun 1993; 16: 251-257
  • 80 Pritchard J, Han R, Horst N, Cruikshank WW, Smith TJ. Immunoglobulin activation of T cell chemoattractant expression in fibroblasts from patients with Graves’ disease is mediated through the insulin-like growth factor 1 receptor pathway. J Immunol 2003; 170: 6348-6354
  • 81 Tsui S, Naik V, Hoa A, Hwang CJ, Afifiyan NF, Sinha AH, Gianoukakis AG, Douglas RS, Smith TJ. Evidence for an Association between Thyroid-Stimulating Hormone and Insulin-Like Growth Factor 1 Receptors: A Tale of Two Antigens Implicated in Graves’ Disease. J Immunol 2008; 181: 4397-4405
  • 82 Varewijck AJ, Boelen A, Lamberts SW, Fliers E, Hofland LJ, Wiersinga WM, Janssen JA. Circulating IgGs may modulate IGF-1 receptor stimulating activity in a subset of patients with Graves’ ophthalmopathy. J Clin Endocrinol Metab 2013; 98: 769-776
  • 83 Minich WB, Dehina N, Welsink T, Schwiebert C, Morgenthaler NG, Kohrle J, Eckstein A, Schomburg L. Autoantibodies to the IGF1 receptor in Graves’ orbitopathy. J Clin Endocrinol Metab 2013; 98: 752-760
  • 84 Gavi S, Shumay E, Wang HY, Malbon CC. G-protein coupled receptors and tyrosine kinases: crossroads in cell signaling and regulation. Trends Endocrinol 2006; 17: 48-54
  • 85 Bartalena L. Graves’ orbitopathy: Imperfect treatments for a rare disease. Eur Thyroid J 2013; 2: 259-269
  • 86 Salvi M, Vannucchi G, Currò N, Campi I, Covelli D, Dazzi D, Simonetta S, Guastella C, Pignataro L, Avignone S, Beck-Peccoz P. Efficacy of B-cell targeted therapy with rituximab in patients with active moderate-severe Graves’ orbitopathy: A randomized controlled study. J Clin Endocrinol Metab 2015; 100: 422-431
  • 87 Stan MN, Garrity JA, Carranza Leon BG, Prabin T, Bradley EA, Bahn R. Randomized controlled trial of rituximab in patients with Graves’ orbitopathy. J Clin Endocrinol Metab 2015; 100: 432-441
  • 88 Nakahara M, Johnson K, Eckstein A, Taguchi R, Yamada M, Abiru N, Nagayama Y. Adoptive transfer of antithyrotropin receptor (TSHR) autoimmunity from TSHR knockout mice to athymic nude mice. Endocrinol 2012; 153: 2034-2042