Ultrasound localization microscopy of renal tumor xenografts in chicken embryo is correlated to hypoxia

Matthew R Lowerison, Chengwu Huang, Fabrice Lucien, Shigao Chen, Pengfei Song, Matthew R Lowerison, Chengwu Huang, Fabrice Lucien, Shigao Chen, Pengfei Song

Abstract

Ultrasound localization microscopy (ULM) permits the reconstruction of super-resolved microvascular images at clinically relevant penetration depths, which can be potentially leveraged to provide non-invasive quantitative measures of tissue hemodynamics and hypoxic status. We demonstrate that ULM microbubble data processing methods, applied to images acquired with a Verasonics Vantage 256 system, can provide a non-invasive imaging surrogate biomarker of tissue oxygenation status. This technique was applied to evaluate the microvascular structure, vascular perfusion, and hypoxia of a renal cell carcinoma xenograft model grown in the chorioallantoic membrane of chicken embryos. Histological microvascular density was significantly correlated to ULM measures of intervessel distance (R = -0.92, CI95 = [-0.99,-0.42], p = 0.01). The Distance Metric, a measure of vascular tortuosity, was found to be significantly correlated to hypoxyprobe quantifications (R = 0.86, CI95 = [0.17, 0.99], p = 0.03). ULM, by providing non-invasive in vivo microvascular structural information, has the potential to be a crucial clinical imaging modality for the diagnosis and therapy monitoring of solid tumors.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Vascular imaging of CAM-engrafted Renca tumors. (A) The ex ovo chicken embryo model has readily accessible surface vasculature, facilitating the injection of precise volumes of microbubble contrast agent. Renca cells engrafted into the CAM surface result in spheroidal tumor masses that are consistently hyper-vascularized. (B) The spheroidal Renca tumors were imaged with ultrasound at five distinct imaging planes to volumetrically sample these renal tumor xenografts. Each imaging plane was imaged using 15 angle plane-wave compounding with a Vantage 256 system (L35-16vX transducer), for a total acquisition length of 3600 frames (7.2 s). (C) B-mode image of example tumor, generated by compounding plane-wave acquisitions, demonstrates a high SNR and well characterized tumor margin. (D) Conventional contrast-enhanced ultrasound image of example tumor confirms the high degree of vascularization typical of CAM-engrafted Renca tumors. Nominal imaging resolution was 50 µm axial and 100 µm lateral. (E) Fluorescent histology section, taken from the example tumor in (D), demonstrates both a high peripheral tumor vascularization (red signal) as well as intratumoral hypoxia (green signal).
Figure 2
Figure 2
Microbubble localization and tracking. (A) IQ data (3600 frames) was represented as a three-dimensional matrix by stacking each B-mode image frame together along a ‘slow-time’ dimension. (B) The resulting imaging volume was reshaped into a 2D Casorati matrix in a column-wise manner, and a singular value decomposition (SVD) was used to remove the highly spatiotemporally coherent tissue background (low singular value orders). (C) An inverse SVD was performed to recover a three-dimensional imaging volume that contained predominantly microbubble signals. A microbubble PSF was convolved with each frame of the dataset shown in (C) to identify the positions of potential microbubble locations. (D) Representative image stack of localized microbubble centroids, after a local maximum search. There are some false positive microbubbles identified in this first pass of localization processing. (E) Bipartite graph-based filtering was used to pair microbubbles along several frames of data. This process reduces the number of false positive microbubble detections by eliminating low-confidence events. (F) Representative microvascular structure map from an example tumor. (G) Blood-flow velocity map for the same representative tumor.
Figure 3
Figure 3
Comparison between diffraction limited CEUS and ULM (A) Contrast-enhanced power images demonstrate that CAM-engraft Renca tumors are highly vascularized through-out the entire tumor volume. (B) Contrast-enhanced color-flow imaging exhibited a mosaic pattern of alternating blood flow velocity directions, with the majority of faster flow typically present in the tumor periphery. (C) Super-resolution microbubble localization maps demonstrated improved resolution of small vessels, while retaining the vascular features of the larger feeding vessels. (D) The microvessel blood-flow velocity images disambiguated some of the velocity information present in the conventional color-flow images, particularly for smaller vessels. The interconnectedness of flow direction is more apparent. (E) Both conventional power images and super-resolution localization maps demonstrated similar trends in their estimate of blood volume for each tumor, with a high significant correlation (R = 0.83, p = 8.7 × 10−7, N = 30, Spearman correlation) (F) Likewise, the correlation between the conventional estimate of FMBV and ULM vascular density was significant, but only moderate in magnitude (R = 0.59, p = 0.0006, N = 30, Spearman correlation) (G) Estimates of mean flow velocity between the two image processing techniques were not found to be significant (R = 0.036, p = 0.85, N = 30, Spearman correlation).
Figure 4
Figure 4
Intratumoral heterogeneity of CAM Renca tumors. (A) Tumor microvessel density maps demonstrate a phenotype of lower microvascular density in the center of the tumor, with most of the large feeding vasculature found in the tumor periphery. (B) ULM blood flow velocity images corroborate these findings, with slower blood flow velocities found in the center of the tumor and a characteristic mosaic patterning of multidirectional blood flow directions. (C) Hypoxyprobe signal demonstrated that the center of this tumor is hypoxic, but showed that the top-most region was well oxygenation (arrow). This may be due to oxygen diffusion from the atmosphere (Supplementary Fig. 1). (D) The tortuosity of the tumor microvasculature was calculated using the Distance Metric and Sum of Angle Metric. (E) Boxplot of the rhodamine MFI, a histological measure of microvascular density. (F) ULM intervessel distance quantifications. (G) Boxplot of hypoxyprobe MFI taken as a quantified measure of intratumoral hypoxia. (H) The Distance Metric for this tumor cohort.
Figure 5
Figure 5
Correlation to fluorescent histology. Correlation matrix of histological measurements in comparison to the vascular metrics derived from ULM imaging (Pearson’s correlation coefficient (R)). Notable findings include a significant negative correlation between rhodamine and ULM intervessel distance (R = −0.92, p = 0.01, N = 6), and a significant positive correlation between hypoxyprobe signal and the Distance Metric (R = 0.86, p = 0.03, N = 6). Significant correlations are denoted with asterisk (*p 

References

    1. Walsh JC, et al. The Clinical Importance of Assessing Tumor Hypoxia: Relationship of Tumor Hypoxia to Prognosis and Therapeutic Opportunities. Antioxid. Redox Signal. 2014;21:1516–1554. doi: 10.1089/ars.2013.5378.
    1. Ferdinand, P. & Roffe, C. Hypoxia after stroke: a review of experimental and clinical evidence. Exp Transl Stroke Med8, (2016).
    1. Roffe C, et al. Effect of Routine Low-Dose Oxygen Supplementation on Death and Disability in Adults With Acute Stroke: The Stroke Oxygen Study Randomized Clinical Trial. JAMA. 2017;318:1125–1135. doi: 10.1001/jama.2017.11463.
    1. Giordano FJ. Oxygen, oxidative stress, hypoxia, and heart failure. J. Clin. Invest. 2005;115:500–508. doi: 10.1172/JCI200524408.
    1. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nat. 2011;473:298–307. doi: 10.1038/nature10144.
    1. Hanahan D, Weinberg RA. Hallmarks of Cancer: The Next Generation. Cell. 2011;144:646–674.
    1. Goel S, et al. Normalization of the vasculature for treatment of cancer and other diseases. Physiol. Rev. 2011;91:1071–1121. doi: 10.1152/physrev.00038.2010.
    1. Cosse J-P, Michiels C. Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression. Anticancer. Agents Med. Chem. 2008;8:790–797. doi: 10.2174/187152008785914798.
    1. Rockwell S, Dobrucki IT, Kim EY, Marrison ST, Vu VT. Hypoxia and radiation therapy: past history, ongoing research, and future promise. Curr. Mol. Med. 2009;9:442–458. doi: 10.2174/156652409788167087.
    1. Barsoum IB, Koti M, Siemens DR, Graham CH. Mechanisms of Hypoxia-Mediated Immune Escape in Cancer. Cancer Res. 2014;74:7185–7190. doi: 10.1158/0008-5472.CAN-14-2598.
    1. Dong Z, Wang JZ, Yu F, Venkatachalam MA. Apoptosis-Resistance of Hypoxic Cells. Am. J. Pathol. 2003;163:663–671. doi: 10.1016/S0002-9440(10)63693-0.
    1. Bristow RG, Hill RP. Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nat. Rev. Cancer. 2008;8:180–192. doi: 10.1038/nrc2344.
    1. Luoto KR, Kumareswaran R, Bristow RG. Tumor hypoxia as a driving force in genetic instability. Genome Integr. 2013;4:5. doi: 10.1186/2041-9414-4-5.
    1. Pennacchietti S, et al. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell. 2003;3:347–361. doi: 10.1016/S1535-6108(03)00085-0.
    1. Vakoc BJ, Fukumura D, Jain RK, Bouma BE. Cancer imaging by optical coherence tomography: preclinical progress and clinical potential. Nat. Rev. Cancer. 2012;12:363–368. doi: 10.1038/nrc3235.
    1. Wang L, Cull G, Cioffi GA. Depth of penetration of scanning laser Doppler flowmetry in the primate optic nerve. Arch. Ophthalmol. 2001;119:1810–1814. doi: 10.1001/archopht.119.12.1810.
    1. Messiou C, et al. Advanced Solid Tumors Treated with Cediranib: Comparison of Dynamic Contrast-enhanced MR Imaging and CT as Markers of Vascular Activity. Radiology. 2012;265:426–436. doi: 10.1148/radiol.12112565.
    1. Zwick S, et al. Simulation-based comparison of two approaches frequently used for dynamic contrast-enhanced MRI. Eur. Radiol. 2010;20:432–442. doi: 10.1007/s00330-009-1556-6.
    1. Hasebroock KM, Serkova NJ. Toxicity of MRI and CT contrast agents. Expert. Opin. Drug. Metab. Toxicol. 2009;5:403–416. doi: 10.1517/17425250902873796.
    1. Dietrich CF, et al. How to perform Contrast-Enhanced Ultrasound (CEUS) Ultrasound Int. Open. 2018;4:E2–E15. doi: 10.1055/s-0043-123931.
    1. Tang M-X, et al. Quantitative contrast-enhanced ultrasound imaging: a review of sources of variability. Interface Focus. 2011;1:520–539. doi: 10.1098/rsfs.2011.0026.
    1. Errico C, et al. Ultrafast ultrasound localization microscopy for deep super-resolution vascular imaging. Nat. 2015;527:499–502. doi: 10.1038/nature16066.
    1. Viessmann OM, Eckersley RJ, Christensen-Jeffries K, Tang MX, Dunsby C. Acoustic super-resolution with ultrasound and microbubbles. Phys. Med. Biol. 2013;58:6447–6458. doi: 10.1088/0031-9155/58/18/6447.
    1. Christensen-Jeffries K, Browning RJ, Tang M-X, Dunsby C, Eckersley RJ. In vivo acoustic super-resolution and super-resolved velocity mapping using microbubbles. IEEE Trans. Med. Imaging. 2015;34:433–440. doi: 10.1109/TMI.2014.2359650.
    1. Couture, O., Besson, B., Montaldo, G., Fink, M. & Tanter, M. Microbubble ultrasound super-localization imaging (MUSLI). in 2011 IEEE International Ultrasonics Symposium 1285–1287, 10.1109/ULTSYM.2011.6293576 (2011).
    1. Lin F, et al. 3-D Ultrasound Localization Microscopy for Identifying Microvascular Morphology Features of Tumor Angiogenesis at a Resolution Beyond the Diffraction Limit of Conventional Ultrasound. Theranostics. 2017;7:196–204. doi: 10.7150/thno.16899.
    1. Bullitt E, Gerig G, Pizer SM, Lin W, Aylward SR. Measuring tortuosity of the intracerebral vasculature from MRA images. IEEE Trans. Med. Imaging. 2003;22:1163–1171. doi: 10.1109/TMI.2003.816964.
    1. Opacic T, et al. Motion model ultrasound localization microscopy for preclinical and clinical multiparametric tumor characterization. Nat. Commun. 2018;9:1527. doi: 10.1038/s41467-018-03973-8.
    1. Dencks S, et al. Clinical Pilot Application of Super-Resolution US Imaging in Breast Cancer. IEEE Trans. Ultrason. Ferroelectr. Freq. Control. 2019;66:517–526. doi: 10.1109/TUFFC.2018.2872067.
    1. Shah A, Bush N, Box G, Eccles S, Bamber J. Value of combining dynamic contrast enhanced ultrasound and optoacoustic tomography for hypoxia imaging. Photoacoust. 2017;8:15–27. doi: 10.1016/j.pacs.2017.08.001.
    1. Huang C, et al. Noninvasive Contrast-Free 3D Evaluation of Tumor Angiogenesis with Ultrasensitive Ultrasound Microvessel Imaging. Sci. Rep. 2019;9:4907. doi: 10.1038/s41598-019-41373-0.
    1. Hingot V, et al. Microvascular flow dictates the compromise between spatial resolution and acquisition time in Ultrasound Localization Microscopy. Sci. Rep. 2019;9:2456. doi: 10.1038/s41598-018-38349-x.
    1. Desailly Y, et al. Contrast enhanced ultrasound by real-time spatiotemporal filtering of ultrafast images. Phys. Med. Biol. 2017;62:31–42. doi: 10.1088/1361-6560/62/1/31.
    1. Song P, et al. Improved Super-Resolution Ultrasound Microvessel Imaging with Spatiotemporal Nonlocal Means Filtering and Bipartite Graph-Based Microbubble Tracking. IEEE Trans. Ultrason. Ferroelectr. Freq. Control. 2018;65:149–167. doi: 10.1109/TUFFC.2017.2778941.
    1. Tang, S. et al. On Kalman Filter–based Microbubble Tracking for Robust Super-Resolution Ultrasound Microvessel Imaging. IEEE transactions on ultrasonics, ferroelectrics, and frequency control(submitted),.
    1. Rubin JM, et al. Fractional moving blood volume: estimation with power Doppler US. Radiology. 1995;197:183–190. doi: 10.1148/radiology.197.1.7568820.
    1. Rubin JM, Bude RO, Carson PL, Bree RL, Adler RS. Power Doppler US: a potentially useful alternative to mean frequency-based color Doppler US. Radiology. 1994;190:853–856. doi: 10.1148/radiology.190.3.8115639.
    1. Shelton SE, et al. Quantification of microvascular tortuosity during tumor evolution utilizing acoustic angiography. Ultrasound Med. Biol. 2015;41:1896–1904. doi: 10.1016/j.ultrasmedbio.2015.02.012.
    1. Rofstad EK, Galappathi K, Mathiesen B, Ruud E-BM. Fluctuating and diffusion-limited hypoxia in hypoxia-induced metastasis. Clin. Cancer Res. 2007;13:1971–1978. doi: 10.1158/1078-0432.CCR-06-1967.
    1. Vaupel P. Hypoxia and Aggressive Tumor Phenotype: Implications for Therapy and Prognosis. Oncologist. 2008;13:21–26. doi: 10.1634/theoncologist.13-S3-21.
    1. Savai R, et al. Evaluation of Angiogenesis Using Micro-Computed Tomography in a Xenograft Mouse Model of Lung Cancer. Neoplasia. 2009;11:48–56. doi: 10.1593/neo.81036.
    1. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Sci. 2005;307:58–62. doi: 10.1126/science.1104819.
    1. Benson DM, Bryan J, Plant AL, Gotto AM, Smith LC. Digital imaging fluorescence microscopy: spatial heterogeneity of photobleaching rate constants in individual cells. J. Cell Biol. 1985;100:1309–1323. doi: 10.1083/jcb.100.4.1309.
    1. Jilani SM, et al. Selective binding of lectins to embryonic chicken vasculature. J. Histochem. Cytochem. 2003;51:597–604. doi: 10.1177/002215540305100505.
    1. Song P, Manduca A, Trzasko JD, Chen S. Ultrasound Small Vessel Imaging With Block-Wise Adaptive Local Clutter Filtering. IEEE Trans. Med. Imaging. 2017;36:251–262. doi: 10.1109/TMI.2016.2605819.
    1. Loupas T, Powers JT, Gill RW. An axial velocity estimator for ultrasound blood flow imaging, based on a full evaluation of the Doppler equation by means of a two-dimensional autocorrelation approach. IEEE Trans. Ultrasonics, Ferroelectrics, Frequency Control. 1995;42:672–688. doi: 10.1109/58.393110.
    1. Hobby JD. Smooth, easy to compute interpolating splines. Discret. Comput. Geom. 1986;1:123–140. doi: 10.1007/BF02187690.
    1. Song P, Manduca A, Trzasko JD, Chen S. Noise Equalization for Ultrafast Plane Wave Microvessel Imaging. IEEE Trans. Ultrason. Ferroelectr. Freq. Control. 2017;64:1776–1781. doi: 10.1109/TUFFC.2017.2748387.
    1. Maibier M, et al. Structure and hemodynamics of vascular networks in the chorioallantoic membrane of the chicken. Am. J. Physiol. Heart Circ. Physiol. 2016;311:H913–H926. doi: 10.1152/ajpheart.00786.2015.
    1. R Core Team. R: A Language and Environment for Statistical Computing. (R Foundation for Statistical Computing 2019).
    1. Wickham, H. ggplot2: Elegant Graphics for Data Analysis. (Springer-Verlag New York, 2016).
    1. Wei, T. & Simko, V. R package ‘corrplot’: Visualization of a Correlation Matrix. (2017).

Source: PubMed

3
Suscribir