Biomarkers for site-specific response to neoadjuvant chemotherapy in epithelial ovarian cancer: relating MRI changes to tumour cell load and necrosis

Jessica M Winfield, Jennifer C Wakefield, James D Brenton, Khalid AbdulJabbar, Antonella Savio, Susan Freeman, Erika Pace, Kerryn Lutchman-Singh, Katherine M Vroobel, Yinyin Yuan, Susana Banerjee, Nuria Porta, Shan E Ahmed Raza, Nandita M deSouza, Jessica M Winfield, Jennifer C Wakefield, James D Brenton, Khalid AbdulJabbar, Antonella Savio, Susan Freeman, Erika Pace, Kerryn Lutchman-Singh, Katherine M Vroobel, Yinyin Yuan, Susana Banerjee, Nuria Porta, Shan E Ahmed Raza, Nandita M deSouza

Abstract

Background: Diffusion-weighted magnetic resonance imaging (DW-MRI) potentially interrogates site-specific response to neoadjuvant chemotherapy (NAC) in epithelial ovarian cancer (EOC).

Methods: Participants with newly diagnosed EOC due for platinum-based chemotherapy and interval debulking surgery were recruited prospectively in a multicentre study (n = 47 participants). Apparent diffusion coefficient (ADC) and solid tumour volume (up to 10 lesions per participant) were obtained from DW-MRI before and after NAC (including double-baseline for repeatability assessment in n = 19). Anatomically matched lesions were analysed after surgical excision (65 lesions obtained from 25 participants). A trained algorithm determined tumour cell fraction, percentage tumour and percentage necrosis on histology. Whole-lesion post-NAC ADC and pre/post-NAC ADC changes were compared with histological metrics (residual tumour/necrosis) for each tumour site (ovarian, omental, peritoneal, lymph node).

Results: Tumour volume reduced at all sites after NAC. ADC increased between pre- and post-NAC measurements. Post-NAC ADC correlated negatively with tumour cell fraction. Pre/post-NAC changes in ADC correlated positively with percentage necrosis. Significant correlations were driven by peritoneal lesions.

Conclusions: Following NAC in EOC, the ADC (measured using DW-MRI) increases differentially at disease sites despite similar tumour shrinkage, making its utility site-specific. After NAC, ADC correlates negatively with tumour cell fraction; change in ADC correlates positively with percentage necrosis.

Clinical trial registration: ClinicalTrials.gov NCT01505829.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Site-specific response of EOC to…
Fig. 1. Site-specific response of EOC to neoadjuvant chemotherapy.
Images in a 62-year-old woman with stage 3 high-grade serous epithelial ovarian cancer show differential response in primary and metastatic lesions: a axial T2-weighted magnetic resonance imaging (MRI) at baseline (pre-NAC), b corresponding axial high-b-value diffusion-weighted MRI (b = 900smm−2), c apparent diffusion coefficient (ADC) map and df matched sections of the same imaging series after three cycles of platinum-based chemotherapy (post-NAC preoperative). (Scalebar on the ADC map is in units of 10−5 mm2 s−1.) Delineation of regions of interest (ROIs) is shown in b, e for the left ovarian lesion (blue ROI) and peritoneal lesion (red ROI). The ovarian lesion remained measurable on MRI after three cycles of chemotherapy and was included in the imaging-pathology comparison, but the peritoneal lesion was nonmeasurable on MRI after three cycles of chemotherapy. MRI magnetic resonance imaging, NAC neoadjuvant chemotherapy, ADC apparent diffusion coefficient, ROI region of interest.
Fig. 2. Probability density functions for ADC…
Fig. 2. Probability density functions for ADC estimates in all lesions at each anatomic site (ovarian, omental and peritoneal lesions and lymph nodes) at baseline (pre-NAC) and after three or four cycles of treatment (post-NAC preoperative).
Probability density functions have been normalised to aid comparison between pre- and post-treatment data. The same points and bandwidth were used for all lesions (bandwidths were determined for each lesion separately and the median bandwidth from all lesions estimated and applied to each lesion). ADC apparent diffusion coefficient, NAC neoadjuvant chemotherapy.
Fig. 3. Deep learning tumour and necrosis…
Fig. 3. Deep learning tumour and necrosis segmentation from pathologist annotations.
a Example of an H&E section from an omental lesion from a 63-year-old woman with stage 3 high-grade serous epithelial ovarian cancer; b shows tumour regions (red) and regions outlined as part of the necrosis (orange) delineated by a pathologist in the lower half of the section. The unannotated standard H&E stain is seen in the top half of the section. The same section after deep learning segmentation of the whole section (c), showing tumour (green) and necrosis (yellow) for comparison. The correlation between the deep learning segmentation and the ground-truth pathologist segmentation is high. The scalebar in a shows 100 microns. H&E haematoxylin and eosin.
Fig. 4. Site-specific correlations between imaging and…
Fig. 4. Site-specific correlations between imaging and histopathology metrics.
Comparison between a preoperative ADCmedian and tumour cell fraction, and b percentage change in ADCmedian and %necrosis, showing all lesions considered together and ovarian, omental, peritoneal lesions and lymph nodes considered separately. r = Spearman correlation coefficient, ADC apparent diffusion coefficient (where ADCmedian is defined as the median ADC of all fitted voxels in a lesion), tumour cell fraction = percentage of viable tumour cells to total cells in sample, %residual tumour = percentage area of whole section represented by viable tumour, %necrosis = percentage area of whole section represented by necrosis.

References

    1. Bergamini A, Candiani M, Taccagni G, Rabaiotti E, Vigano R, De Marzi P, et al. Different patterns of disease spread between advanced-stage type I and II epithelial ovarian cancer. Gynecol. Obstet. Invest. 2016;81:10–14. doi: 10.1159/000381261.
    1. Horowitz NS, Miller A, Rungruang B, Richard SD, Rodriguez N, Bookman MA, et al. Does aggressive surgery improve outcomes? Interaction between preoperative disease burden and complex surgery in patients with advanced-stage ovarian cancer: an analysis of GOG 182. J. Clin. Oncol. 2015;33:937–943. doi: 10.1200/JCO.2014.56.3106.
    1. Vergote I, Trope CG, Amant F, Kristensen GB, Ehlen T, Johnson N, et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N. Engl. J. Med. 2010;363:943–953. doi: 10.1056/NEJMoa0908806.
    1. Kyriazi S, Nye E, Stamp G, Collins DJ, Kaye SB, deSouza NM. Value of diffusion-weighted imaging for assessing site-specific response of advanced ovarian cancer to neoadjuvant chemotherapy: correlation of apparent diffusion coefficients with epithelial and stromal densities on histology. Cancer Biomark. 2010;7:201–210. doi: 10.3233/CBM-2010-0194.
    1. Sala E, Kataoka MY, Priest AN, Gill AB, McLean MA, Joubert I, et al. Advanced ovarian cancer: multiparametric MR imaging demonstrates response- and metastasis-specific effects. Radiology. 2012;263:149–159. doi: 10.1148/radiol.11110175.
    1. Miow QH, Tan TZ, Ye J, Lau JA, Yokomizo T, Thiery JP, et al. Epithelial-mesenchymal status renders differential responses to cisplatin in ovarian cancer. Oncogene. 2015;34:1899–1907. doi: 10.1038/onc.2014.136.
    1. Burger RA, Brady MF, Rhee J, Sovak MA, Kong G, Nguyen HP, et al. Independent radiologic review of the Gynecologic Oncology Group Study 0218, a phase III trial of bevacizumab in the primary treatment of advanced epithelial ovarian, primary peritoneal, or fallopian tube cancer. Gynecol. Oncol. 2013;131:21–26. doi: 10.1016/j.ygyno.2013.07.100.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur. J. Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Husain A, Wang Y, Hanker LC, Ojeda B, Anttila M, Breda E, et al. Independent radiologic review of AURELIA, a phase 3 trial of bevacizumab plus chemotherapy for platinum-resistant recurrent ovarian cancer. Gynecol. Oncol. 2016;142:465–470. doi: 10.1016/j.ygyno.2016.05.011.
    1. Abramson RG, Arlinghaus LR, Dula AN, Quarles CC, Stokes AM, Weis JA, et al. MR imaging biomarkers in oncology clinical trials. Magn. Reson. Imaging Clin. N. Am. 2016;24:11–29. doi: 10.1016/j.mric.2015.08.002.
    1. de Perrot T, Lenoir V, Domingo Ayllon M, Dulguerov N, Pusztaszeri M, Becker M. Apparent diffusion coefficient histograms of human papillomavirus-positive and human papillomavirus-negative head and neck squamous cell carcinoma: assessment of tumor heterogeneity and comparison with histopathology. Am. J. Neuroradiol. 2017;38:2153–2160. doi: 10.3174/ajnr.A5370.
    1. Rosenkrantz AB, Sigmund EE, Winnick A, Niver BE, Spieler B, Morgan GR, et al. Assessment of hepatocellular carcinoma using apparent diffusion coefficient and diffusion kurtosis indices: preliminary experience in fresh liver explants. Magn. Reson. Imaging. 2012;30:1534–1540. doi: 10.1016/j.mri.2012.04.020.
    1. Arunachalam, H. B., Mishra, R., Daescu, O., Cederberg, K., Rakheja, D., Sengupta, A. et al. Viable and necrotic tumor assessment from whole slide images of osteosarcoma using machine-learning and deep-learning models. PLoS ONE14, e0210706 (2019).
    1. Winfield JM, Wakefield JC, Dolling D, Hall M, Freeman S, Brenton JD, et al. Diffusion-weighted MRI in advanced epithelial ovarian cancer: apparent diffusion coefficient as a response marker. Radiology. 2019;293:374–383. doi: 10.1148/radiol.2019190545.
    1. Winfield JM, Collins DJ, Priest AN, Quest RA, Glover A, Hunter S, et al. A framework for optimization of diffusion-weighted MRI protocols for large field-of-view abdominal-pelvic imaging in multicenter studies. Med. Phys. 2016;43:95. doi: 10.1118/1.4937789.
    1. Blackledge MD, Leach MO, Collins DJ, Koh DM. Computed diffusion-weighted MR imaging may improve tumor detection. Radiology. 2011;261:573–581. doi: 10.1148/radiol.11101919.
    1. AbdulJabbar K, Raza SEA, Rosenthal R, Jamal-Hanjani M, Veeriah S, Akarca A, et al. Geospatial immune variability illuminates differential evolution of lung adenocarcinoma. Nat. Med. 2020;26:1054–1062. doi: 10.1038/s41591-020-0900-x.
    1. Raza SEA, Cheung L, Shaban M, Graham S, Epstein D, Pelengaris S, et al. Micro-Net: a unified model for segmentation of various objects in microscopy images. Med. Image Anal. 2019;52:160–173. doi: 10.1016/j.media.2018.12.003.
    1. Bland JM, Altman DG. Statistical methods for assessing agreement between two methods of clinical measurement. Lancet. 1986;327:307–310. doi: 10.1016/S0140-6736(86)90837-8.
    1. Langer DL, van der Kwast TH, Evans AJ, Plotkin A, Trachtenberg J, Wilson BC, et al. Prostate tissue composition and MR measurements: investigating the relationships between ADC, T2, K trans, ve, and corresponding histologic features. Radiology. 2010;255:485–494. doi: 10.1148/radiol.10091343.
    1. Yuan SJ, Qiao TK, Qiang JW. Diffusion-weighted imaging and diffusion kurtosis imaging for early evaluation of the response to docetaxel in rat epithelial ovarian cancer. J. Transl. Med. 2018;16:340. doi: 10.1186/s12967-018-1714-1.
    1. Fu C, Feng X, Bian D, Zhao Y, Fang X, Du W, et al. Simultaneous changes of magnetic resonance diffusion-weighted imaging and pathological microstructure in locally advanced cervical cancer caused by neoadjuvant chemotherapy. J. Magn. Reson Imaging. 2015;42:427–435. doi: 10.1002/jmri.24779.
    1. Yin Y, Sedlaczek O, Muller B, Warth A, Gonzalez-Vallinas M, Lahrmann B, et al. Tumor cell load and heterogeneity estimation from diffusion-weighted MRI calibrated with histological data: an example from lung cancer. IEEE Trans. Med. Imaging. 2018;37:35–46. doi: 10.1109/TMI.2017.2698525.
    1. Klau M, Gaida MM, Lemke A, Grünberg K, Simon D, Wente MN, et al. Fibrosis and pancreatic lesions: counterintuitive behavior of the diffusion imaging–derived structural diffusion coefficient D. Invest. Radiol. 2013;48:129–133. doi: 10.1097/RLI.0b013e31827ac0f1.
    1. Yamaguchi K, Hara Y, Kitano I, Hamamoto T, Kiyomatsu K, Yamasaki F, et al. Tumor-stromal ratio (TSR) of invasive breast cancer: correlation with multi-parametric breast MRI findings. Br. J. Radiol. 2019;92:20181032. doi: 10.1259/bjr.20181032.
    1. Messiou C, Giles S, Collins DJ, West S, Davies FE, Morgan GJ, et al. Assessing response of myeloma bone disease with diffusion-weighted MRI. Br. J. Radiol. 2012;85:e1198–e1203. doi: 10.1259/bjr/52759767.
    1. Albano D, Patti C, Matranga D, Lagalla R, Midiri M, Galia M. Whole-body diffusion-weighted MR and FDG-PET/CT in Hodgkin Lymphoma: predictive role before treatment and early assessment after two courses of ABVD. Eur. J. Radiol. 2018;103:90–98. doi: 10.1016/j.ejrad.2018.04.014.
    1. Hagtvedt T, Seierstad T, Lund KV, Londalen AM, Bogsrud TV, Smith HJ, et al. Diffusion-weighted MRI compared to FDG PET/CT for assessment of early treatment response in lymphoma. Acta Radiol. 2015;56:152–158. doi: 10.1177/0284185114526087.
    1. Scalco E, Marzi S, Sanguineti G, Vidiri A, Rizzo G. Characterization of cervical lymph-nodes using a multi-parametric and multi-modal approach for an early prediction of tumor response to chemo-radiotherapy. Phys. Med. 2016;32:1672–1680. doi: 10.1016/j.ejmp.2016.09.003.
    1. Weiss E, Ford JC, Olsen KM, Karki K, Saraiya S, Groves R, et al. Apparent diffusion coefficient (ADC) change on repeated diffusion-weighted magnetic resonance imaging during radiochemotherapy for non-small cell lung cancer: a pilot study. Lung Cancer. 2016;96:113–119. doi: 10.1016/j.lungcan.2016.04.001.
    1. Min M, Lee MT, Lin P, Holloway L, Wijesekera D, Gooneratne D, et al. Assessment of serial multi-parametric functional MRI (diffusion-weighted imaging and R 2*) with 18F-FDG-PET in patients with head and neck cancer treated with radiation therapy. Br. J. Radiol. 2016;89:20150530. doi: 10.1259/bjr.20150530.
    1. Carlin D, Weller A, Kramer G, Liu Y, Waterton JC, Chiti A, et al. Evaluation of diffusion-weighted MRI and (18F) fluorothymidine-PET biomarkers for early response assessment in patients with operable non-small cell lung cancer treated with neoadjuvant chemotherapy. BJR Open. 2019;1:20190029.
    1. Wang J, Sun M, Liu D, Hu X, Pui MH, Meng Q, et al. Correlation between apparent diffusion coefficient and histopathology subtypes of osteosarcoma after neoadjuvant chemotherapy. Acta Radiol. 2017;58:971–976. doi: 10.1177/0284185116678276.
    1. Weigelt B, Vargas HA, Selenica P, Geyer FC, Mazaheri Y, Blecua P, et al. Radiogenomics analysis of intratumor heterogeneity in a patient with high-grade serous ovarian cancer. JCO Precis. Oncol. 2019;3:1–9.

Source: PubMed

3
Subscribe