AccScience Publishing / TD / Volume 2 / Issue 3 / DOI: 10.36922/td.1414
Cite this article
132
Download
751
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
ORIGINAL RESEARCH ARTICLE

Early results in the novel use of contrast-enhanced susceptibility-weighted imaging in the assessment of response and progression in desmoid fibromatosis: A pilot study in a specialized cancer institution

Raul F. Valenzuela1†* Elvis Duran Sierra1† Mathew A. Canjirathinkal1 Colleen M. Costelloe1 John E. Madewell1 William A. Murphy Jr.1 Behrang Amini1
Show Less
1 Department of Musculoskeletal Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
Tumor Discovery 2023, 2(3), 1414 https://doi.org/10.36922/td.1414
Submitted: 30 July 2023 | Accepted: 10 October 2023 | Published: 6 November 2023
© 2023 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Routine radiologic reporting (RRR) often considers progressive desmoid tumors to have a higher proportion of T2-hyperintense and T1-shortened-enhancing components, while responsive or mature collagenized tumors demonstrate a higher proportion of T2-hypointense-non-enhancing components. We aim to determine the utility of the novel use of contrast-enhanced susceptibility-weighted imaging (CE-SWI) in Desmoid-Tumor treatment response assessment, distinguishing between the T1-shortening-enhancing/T2-hyperintense immature components from the T2-hypointense mature collagenized components. This pilot study included 10 single-lesion extremity desmoid fibromatosis patients undergoing standard-of-care magnetic resonance imaging, including CE-SWI. Three-dimensional (3D) tumor segmentation was performed using MIM software in 48 volumes of interest. Maximum diameter, volume, and modified Choi (mChoi) measurements were computed from CE-SWI and T2-weighted image (T2-WI). Five first-order radiomic features, including mean, skewness, kurtosis, and 10th and 90th percentiles, were calculated using in-house developed software (CARPI-AF). (i) RECIST Progression: We observed two cases of progression according to the T2-WI-based Response Evaluation Criteria in Solid Tumors standard (RECIST). Interestingly, CE-SWI-based-volume and CE-SWI-based-mChoi predicted the same assessment 4.5 months earlier than T2-WI-based-RECIST. RRR assessed both cases as progression; (ii) RECIST Stability: Out of the eight patients classified as having stable disease by T2-WI-based-RECIST, four discrepant progressions were determined: three patients showed an increase greater than 25% of T2-WI-based-volume, and two patients showed an increase greater than 25% of CE-SWI-based-volume. Moreover, from the RECIST stable group, four discrepant-positive responses were predicted by CE-SWI-based-mChoi (three patients) and T2-WI-based-mChoi (four patients). RRR only assessed one patient as having progressive disease; (iii) First-Order Radiomics: CE-SWI detected 23% more 90th-percentile voxels than T2-WI, while T2-WI demonstrated 8.5% more 10th-percentile voxels than CE-SWI. Notably, expected first-order response/progression-related changes in 10th-percentile, 90th-percentile, mean, and skewness were present in 90% of cases. In conclusion, CE-SWI-based-volume and CE-SWI-based-mChoi measurements could improve the prediction of response/progression in desmoid tumors, enhancing the ability in discriminating between T2*- hypointense-collagenized-mature and T1-shortened-enhancing immature components, respectively, in predominant mature responsive and immature progressive tumors, respectively. RRR is relatively insensitive to volumetric tumor changes before RECIST progression and tends to be better tuned with T2* signal and enhancement changes.

Keywords
Susceptibility weighted imaging
Desmoid fibromatosis
First order radiomics
Modified-Choi
Funding
M.R. Evelyn Hudson Foundation Endowed Professorship
John S. Dunn, Sr. Distinguished Chair in Diagnostic Imaging
References
  1. Fletcher CD, Unni K, Mertens F, 2002, World Health Organization classification of tumours. In: Pathology and Genetics of Tumours of Soft Tissue and Bone. Lyon, France: IARC press.

 

  1. Crombé A, Kind M, Ray-Coquard I, et al., 2020, Progressive desmoid tumor: Radiomics compared with conventional response criteria for predicting progression during systemic therapy-a multicenter study by the French sarcoma group. AJR Am J Roentgenol, 215(6): 1539–1548. https://doi.org/10.2214/AJR.19.22635

 

  1. Wu C, Amini-Nik S, Nadesan P, et al., 2010, Aggressive fibromatosis (desmoid tumor) is derived from mesenchymal progenitor cells. Cancer Res, 70(19): 7690–7698. https://doi.org/10.1158/0008-5472.CAN-10-1656

 

  1. Li M, Cordon-Cardo C, Gerald WL, et al., 1996, Desmoid fibromatosis is a clonal process. Hum Pathol, 27(9): 939–943. https://doi.org/10.1016/s0046-8177(96)90221-x

 

  1. Cadour F, Tradi F, Bartoli A, et al., 2023, Diffusion weighted imaging changes in extra-abdominal desmoid tumor after cryotherapy. Ann Med, 55(1): 521–525. https://doi.org/10.1080/07853890.2023.2174589

 

  1. Sheth PJ, Del Moral S, Wilky BA, et al., 2016, Desmoid fibromatosis: MRI features of response to systemic therapy. Skeletal Radiol, 45: 1365–1373. https://doi.org/10.1007/s00256-016-2439-y

 

  1. Shinagare AB, Ramaiya NH, Jagannathan JP, et al., 2011, A to Z of desmoid tumors. AJR Am J Roentgenol, 197(6): W1008–W1014. https://doi.org/10.2214/AJR.11.6657

 

  1. Sundaram M, McGuire MH, Schajowicz F, 1987, Soft-tissue masses: Histologic basis for decreased signal (short T2) on T2-weighted MR images. AJR Am J Roentgenol, 148(6): 1247–1250. https://doi.org/10.2214/ajr.148.6.1247

 

  1. Gounder MM, Lefkowitz RA, Keohan ML, et al., 2011, Activity of Sorafenib against desmoid tumor/deep fibromatosis. Clin Cancer Res, 17(12): 4082–4090. https://doi.org/10.1158/1078-0432.ccr-10-3322

 

  1. Zanchetta E, Ciniselli C, Bardelli A, et al., 2021, Magnetic resonance imaging patterns of tumor response to chemotherapy in desmoid‐type fibromatosis. Cancer Med, 10(13): 4356–4365. https://doi.org/10.1002/cam4.3973

 

  1. Subhawong TK, Feister K, Sweet K, et al., 2021, MRI volumetrics and image texture analysis in assessing systemic treatment response in extra-abdominal desmoid fibromatosis. Radiol Imaging Cancer, 3(4): e210016. https://doi.org/10.1148/rycan.2021210016

 

  1. Zhou MY, Bui NQ, Charville GW, et al., 2022, Current management and recent progress in desmoid tumors. Cancer Treat Res Commun, 31: 100562. https://doi.org/10.1016/j.ctarc.2022.100562

 

  1. Otero S, Moskovic EC, Strauss DC, et al., 2015, Desmoid-type fibromatosis. Clin Radiol, 70(9): 1038–1045. https://doi.org/10.1016/j.crad.2015.04.015

 

  1. Cassidy MR, Lefkowitz RA, Long N, et al., 2020, Association of MRI T2 signal intensity with desmoid tumor progression during active observation: A retrospective cohort study. Ann Surg, 271: 748–755. https://doi.org/10.1097/SLA.0000000000003073

 

  1. Valenzuela RF, Madewell JE, Kundra V, et al., 2021, Advanced imaging in musculoskeletal oncology: Moving away from RECIST and embracing advanced bone and soft tissue tumor imaging (ABASTI)-part II-novel functional imaging techniques. Semin Ultrasound CT MR, 42(2): 215–227. https://doi.org/10.1053/j.sult.2020.08.013

 

  1. Haacke EM, Reichenbach JR, 2014, Susceptibility Weighted Imaging in MRI: Basic Concepts and Clinical Applications. Hoboken: John Wiley and Sons.

 

  1. Halefoglu AM, Yousem DM, 2018, Susceptibility weighted imaging: Clinical applications and future directions. World J Radiol, 10(4): 30–45. https://doi.org/10.4329/wjr.v10.i4.30

 

  1. Haller S, Haacke EM, Thurnher MM, et al., 2021, Susceptibility-weighted imaging: Technical essentials and clinical neurologic applications. Radiology, 299(1): 3–26. https://doi.org/10.1148/radiol.2021203071

 

  1. Skalski KA, Kessler AT, Bhatt AA, 2018, Hemorrhagic and non-hemorrhagic causes of signal loss on susceptibility-weighted imaging. Emerg Radiol, 25: 691–701. https://doi.org/10.1007/s10140-018-1634-7

 

  1. Eisenhauer EA, Therasse P, Bogaerts J, et al., 2009, New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur J Cancer, 45(2): 228–247. https://doi.org/10.1016/j.ejca.2008.10.026

 

  1. Therasse P, Arbuck SG, Eisenhauer EA, et al., 2000, New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst, 92(3): 205–216. https://doi.org/10.1093/jnci/92.3.205

 

  1. Ko CC, Yeh LR, Kuo YT, et al., 2021, Imaging biomarkers for evaluating tumor response: RECIST and beyond. Biomarker Res, 9(1): 52. https://doi.org/10.1186/s40364-021-00306-8

 

  1. Koh DM, Collins DJ, 2007, Diffusion-weighted MRI in the body: Applications and challenges in oncology. AJR Am J Roentgenol, 188(6): 1622–1635. https://doi.org/10.2214/AJR.06.1403

 

  1. Messiou C, Collins DJ, Morgan VA, et al., 2014, Use of apparent diffusion coefficient as a response biomarker in bone: Effect of developing sclerosis on quantified values. Skeletal Radiol, 43(2): 205–208. https://doi.org/10.1007/s00256-013-1768-3

 

  1. Jahng GH, Li KL, Ostergaard L, et al., 2014, Perfusion magnetic resonance imaging: A comprehensive update on principles and techniques. Korean J Radiol, 15(5): 554–577. https://doi.org/10.3348/kjr.2014.15.5.554

 

  1. Essig M, Shiroishi MS, Nguyen TB, et al., 2013, Perfusion MRI: The five most frequently asked technical questions. AJR Am J Roentgenol, 200(1): 24–34. https://doi.org/10.2214/AJR.12.9543

 

  1. Stacchiotti S, Collini P, Messina A, et al., 2009, High-grade soft-tissue sarcomas: Tumor response assessment--pilot study to assess the correlation between radiologic and pathologic response by using RECIST and Choi criteria. Radiology, 251(2): 447–456. https://doi.org/10.1148/radiol.2512081403

 

  1. Zwanenburg A, Vallières M, Abdalah MA, et al., 2020, The image biomarker standardization initiative: Standardized quantitative radiomics for high-throughput image-based phenotyping. Radiology, 295(2):328-338. https://doi.org/10.1148/radiol.2020191145

 

  1. Vallières M, Zwanenburg A, Badic B, et al., 2018, Responsible radiomics research for faster clinical translation. J Nucl Med, 59(2):189-193. https://doi.org/10.2967/jnumed.117.200501

 

  1. Van Timmeren JE, Cester D, Tanadini-Lang S, et al., 2020, Radiomics in medical imaging-“how-to” guide and critical reflection. Insights Imaging, 11(1): 91. https://doi.org/10.1186/s13244-020-00887-2

 

  1. Shah GD, Kesari S, Xu R, et al., 2006, Comparison of linear and volumetric criteria in assessing tumor response in adult high-grade gliomas. Neuro Oncol, 8(1): 38–46. https://doi.org/10.1215/S1522851705000529
Conflict of interest
The authors declare that they have no competing interests.
Share
Back to top
Tumor Discovery, Electronic ISSN: 2810-9775 Published by AccScience Publishing