DOI QR코드

DOI QR Code

T2 Mapping with and without Fat-Suppression to Predict Treatment Response to Intravenous Glucocorticoid Therapy for Thyroid-Associated Ophthalmopathy

  • Linhan Zhai (Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology) ;
  • Qiuxia Wang (Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology) ;
  • Ping Liu (Department of Medical Imaging, Guangdong Second Provincial General Hospital) ;
  • Ban Luo (Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology) ;
  • Gang Yuan (Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology) ;
  • Jing Zhang (Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology)
  • Received : 2021.08.09
  • Accepted : 2022.03.15
  • Published : 2022.06.01

Abstract

Objective: To evaluate the performance of baseline clinical characteristics and pretherapeutic histogram parameters derived from T2 mapping of the extraocular muscles (EOMs) in the prediction of treatment response to intravenous glucocorticoid (IVGC) therapy for active and moderate-to-severe thyroid-associated ophthalmopathy (TAO) and to investigate the effect of fat-suppression (FS) in T2 mapping in this prediction. Materials and Methods: A total of 79 patients clinically diagnosed with active, moderate-to-severe TAO (47 female, 32 male; mean age ± standard deviation, 46.1 ± 10 years), including 43 patients with a total of 86 orbits in the responsive group and 36 patients with a total of 72 orbits in the unresponsive group, were enrolled. Baseline clinical characteristics and pretherapeutic histogram parameters derived from T2 mapping with FS (i.e., FS T2 mapping) or without FS (i.e., conventional T2 mapping) of EOMs were compared between the two groups. Independent predictors of treatment response to IVGC were identified using multivariable analysis. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive performance of the prediction models. Differences between the models were examined using the DeLong test. Results: Compared to the unresponsive group, the responsive group had a shorter disease duration, lower kurtosis (FS-kurtosis), lower standard deviation, larger 75th, 90th, and 95th (FS-95th) T2 relaxation times in FS mapping and lower kurtosis in conventional T2 mapping. Multivariable analysis revealed that disease duration, FS-95th percentile, and FS-kurtosis were independent predictors of treatment response. The combined model, integrating all identified predictors, had an optimized area under the ROC curve of 0.797, 88.4% sensitivity, and 62.5% specificity, which were significantly superior to those of the imaging model (p = 0.013). Conclusion: An integrated combination of disease duration, FS-95th percentile, and FS-kurtosis was a potential predictor of treatment response to IVGC in patients with active and moderate-to-severe TAO. FS T2 mapping was superior to conventional T2 mapping in terms of prediction.

Keywords

Acknowledgement

We would like to thank Editage (www.editage.cn) for English language editing.

References

  1. Bahn RS. Graves' ophthalmopathy. N Engl J Med 2010;362:726-738
  2. Bartalena L, Baldeschi L, Boboridis K, Eckstein A, Kahaly GJ, Marcocci C, et al. The 2016 European Thyroid Association/European Group on Graves' orbitopathy guidelines for the management of Graves' orbitopathy. Eur Thyroid J 2016;5:9-26
  3. Vannucchi G, Covelli D, Campi I, Origo D, Curro N, Cirello V, et al. The therapeutic outcome to intravenous steroid therapy for active Graves' orbitopathy is influenced by the time of response but not polymorphisms of the glucocorticoid receptor. Eur J Endocrinol 2014;170:55-61
  4. Sussman MS, Vidarsson L, Pauly JM, Cheng HL. A technique for rapid single-echo spin-echo T2 mapping. Magn Reson Med 2010;64:536-545
  5. Keene KR, Beenakker JM, Hooijmans MT, Naarding KJ, Niks EH, Otto LAM, et al. T2 relaxation-time mapping in healthy and diseased skeletal muscle using extended phase graph algorithms. Magn Reson Med 2020;84:2656-2670
  6. Chen W, Hu H, Chen HH, Su GY, Yang T, Xu XQ, et al. Utility of T2 mapping in the staging of thyroid-associated ophthalmopathy: efficiency of region of interest selection methods. Acta Radiol 2020;61:1512-1519
  7. Das T, Roos JCP, Patterson AJ, Graves MJ, Murthy R. T2-relaxation mapping and fat fraction assessment to objectively quantify clinical activity in thyroid eye disease: an initial feasibility study. Eye (Lond) 2019;33:235-243
  8. Hu H, Xu XQ, Chen L, Chen W, Wu Q, Chen HH, et al. Predicting the response to glucocorticoid therapy in thyroid-associated ophthalmopathy: mobilizing structural MRI-based quantitative measurements of orbital tissues. Endocrine 2020;70:372-379
  9. Shafi F, Mathewson P, Mehta P, Ahluwalia HS. The enlarged extraocular muscle: to relax, reflect or refer? Eye (Lond) 2017;31:537-544
  10. Gould DJ, Roth FS, Soparkar CN. The diagnosis and treatment of thyroid-associated ophthalmopathy. Aesthetic Plast Surg 2011;36:638-648
  11. Zhang C, Lin Y, Han Z, Gao L, Guo R, Shi Q, et al. Feasibility of T2 mapping and magnetic transfer ratio for diagnosis of intervertebral disc degeneration at the cervicothoracic junction: a pilot study. Biomed Res Int 2019;2019:6396073
  12. Del Grande F, Santini F, Herzka DA, Aro MR, Dean CW, Gold GE, et al. Fat-suppression techniques for 3-T MR imaging of the musculoskeletal system. Radiographics 2014;34:217-233
  13. Lubner MG, Smith AD, Sandrasegaran K, Sahani DV, Pickhardt PJ. CT texture analysis: definitions, applications, biologic correlates, and challenges. Radiographics 2017;37:1483-1503
  14. Hou K, Ai T, Hu WK, Luo B, Wu YP, Liu R. Three dimensional orbital magnetic resonance T2-mapping in the evaluation of patients with Graves' ophthalmopathy. J Huazhong Univ Sci Technolog Med Sci 2017;37:938-942
  15. Mourits MP, Prummel MF, Wiersinga WM, Koornneef L. Clinical activity score as a guide in the management of patients with Graves' ophthalmopathy. Clin Endocrinol (Oxf) 1997;47:9-14
  16. Bartalena L, Baldeschi L, Dickinson A, Eckstein A, Kendall-Taylor P, Marcocci C, et al. Consensus statement of the European Group on Graves' orbitopathy (EUGOGO) on management of GO. Eur J Endocrinol 2008;158:273-285
  17. Xu L, Li L, Xie C, Guan M, Xue Y. Thickness of extraocular muscle and orbital fat in MRI predicts response to glucocorticoid therapy in Graves' ophthalmopathy. Int J Endocrinol 2017;2017:3196059
  18. Shen L, Huang F, Ye L, Zhu W, Zhang X, Wang S, et al. Circulating microRNA predicts insensitivity to glucocorticoid therapy in Graves' ophthalmopathy. Endocrine 2015;49:445-456
  19. Prabhakar BS, Bahn RS, Smith TJ. Current perspective on the pathogenesis of Graves' disease and ophthalmopathy. Endocr Rev 2003;24:802-835
  20. Smith TJ, Koumas L, Gagnon A, Bell A, Sempowski GD, Phipps RP, et al. Orbital fibroblast heterogeneity may determine the clinical presentation of thyroid-associated ophthalmopathy. J Clin Endocrinol Metab 2002;87:385-392
  21. Ohnishi T, Noguchi S, Murakami N, Tajiri J, Harao M, Kawamoto H, et al. Extraocular muscles in Graves ophthalmopathy: usefulness of T2 relaxation time measurements. Radiology 1994;190:857-862
  22. Gold GE, Han E, Stainsby J, Wright G, Brittain J, Beaulieu C. Musculoskeletal MRI at 3.0 T: relaxation times and image contrast. AJR Am J Roentgenol 2004;183:343-351
  23. Schlaeger S, Weidlich D, Klupp E, Montagnese F, Deschauer M, Schoser B, et al. Water T2 mapping in fatty infiltrated thigh muscles of patients with neuromuscular diseases using a T2-prepared 3D turbo spin echo with SPAIR. J Magn Reson Imaging 2020;51:1727-1736
  24. Bednarczuk T, Gopinath B, Ploski R, Wall JR. Susceptibility genes in Graves' ophthalmopathy: searching for a needle in a haystack? Clin Endocrinol (Oxf) 2007;67:3-19
  25. Lacheta D, Mis' kiewicz P, Gluszko A, Nowicka G, Struga M, Kantor I, et al. Immunological aspects of Graves' ophthalmopathy. Biomed Res Int 2019;2019:7453260
  26. Wang Y, Zhang S, Zhang Y, Liu X, Gu H, Zhong S, et al. A single-center retrospective study of factors related to the effects of intravenous glucocorticoid therapy in moderate-to-severe and active thyroid-associated ophthalmopathy. BMC Endocr Disord 2018;18:13
  27. S,ahli E, Gunduz K. Thyroid-associated Ophthalmopathy. Turk J Ophthalmol 2017;47:94-105
  28. Xing L, Ye L, Zhu W, Shen L, Huang F, Jiao Q, et al. Smoking was associated with poor response to intravenous steroids therapy in Graves' ophthalmopathy. Br J Ophthalmol 2015;99:1686-1691
  29. Wiersinga WM. Advances in treatment of active, moderate-to-severe Graves' ophthalmopathy. Lancet Diabetes Endocrinol 2017;5:134-142
  30. Liu P, Luo B, Chen L, Wang QX, Yuan G, Jiang GH, et al. Baseline volumetric T2 relaxation time histogram analysis: can it be used to predict the response to intravenous methylprednisolone therapy in patients with thyroid-associated ophthalmopathy? Front Endocrinol (Lausanne) 2021;12:614536
  31. Hu H, Chen HH, Chen W, Wu Q, Chen L, Zhu H, et al. T2 mapping histogram at extraocular muscles for predicting the response to glucocorticoid therapy in patients with thyroid-associated ophthalmopathy. Clin Radiol 2021;76:159.e1-159. e8