Characterization of Anti-Angiogenic Chemo-Sensitization via Longitudinal Ultrasound Localization Microscopy in Colorectal Carcinoma Tumor Xenografts

Matthew Lowerison, Wei Zhang, Xi Chen, Timothy Fan, Pengfei Song, Matthew Lowerison, Wei Zhang, Xi Chen, Timothy Fan, Pengfei Song

Abstract

Objective: Super-resolution ultrasound localization microscopy (ULM) has unprecedented vascular resolution at clinically relevant imaging penetration depths. This technology can potentially screen for the transient microvascular changes that are thought to be critical to the synergistic effect(s) of combined chemotherapy-antiangiogenic agent regimens for cancer.

Methods: In this paper, we apply this technology to a high-throughput colorectal carcinoma xenograft model treated with either the antiangiogenic agent sorafenib, FOLFOX-6 chemotherapy, a combination of the two treatments, or vehicle control.

Results: Longitudinal ULM demonstrated morphological changes in the antiangiogenic treated cohorts, and evidence of vascular disruption caused by chemotherapy. Gold-standard histological measurements revealed reduced levels of hypoxia in the sorafenib treated cohort for both of the human cell lines tested (HCT-116 and HT-29). Therapy resistance was associated with an increase in tumor vascular fractal dimension as measured by a box-counting technique on ULM images.

Conclusion: These results imply that the morphological changes evident on ULM signify a functional change in the tumor microvasculature, which may be indicative of chemo-sensitivity.

Significance: ULM provides additional utility for tumor therapy response evaluation by offering a myriad of morphological and functional quantitative indices for gauging treatment effect(s).

Figures

Fig. 1.
Fig. 1.
Study design diagram. A) At EDD-09 a total of 52 CAMs were engrafted with the HCT-116 cell line and 47 CAMs were engrafted with the HT-29 cell line. B) The tumor engrafted CAMs were randomized, and a pretreatment ULM imaging session was performed on EDD-13. C) The tumors underwent their selected therapy as described in the methods section, and a post-treatment ULM imaging session was performed on EDD-17. Tumors were excised immediately after imaging and formalin fixed for histology.
Fig. 2.
Fig. 2.
ULM acquisition and processing pipeline. A) A reference transducer acquisition was used to estimate a noise equalization profile to correct for depth dependent attenuation. B) A total of 32000 contrast-enhanced IQ data frames were acquired and SVD filtered to generate isolated microbubble data and C) contrast-enhanced power Doppler images. The isolated microbubble data then went into the ULM processing pipeline, which included microbubble separation, microbubble localization, and microbubble pairing and tracking. D) The final super-resolved images were used to quantify the treatment effect in each CAM tumor therapy group.
Fig. 3.
Fig. 3.
Longitudinal changes in CAM tumors. A) Representative baseline contrast-enhanced power Doppler and super-resolution ULM images of CAM engrafted tumors. B) This same control tumor at endpoint imaging, which demonstrates both the growth of the tumor diameter and the continued development of intratumor vascularization over the course of the study. Tumor outlines and maximal diameter are demonstrated as cyan and orange lines on the B-mode image, respectively. C) Quantification of the tumor long-axis which reveals a general trend of tumor growth for all treatment groups in this study. D) Quantification of the contrast power of the tumor cross-sectional area.
Fig. 4.
Fig. 4.
Super-resolution images of treatment effect. A) Endpoint (EDD-17) control ULM images reveal a dense microvascular network throughout the tumor mass. B) In comparison to the control tumors, the sorafenib treated tumors exhibited slight vascular pruning of microvessels, with a more columnar appearance in vascular network structure (arrow). C) Chemotherapy treated tumors displayed avascular tumor regions, implying a more aggressive vascular pruning. D) Combination treated tumors also exhibit avascular tumor regions.
Fig. 5.
Fig. 5.
Histological comparison and quantifications. A) The super-resolution vascular map can be compared qualitatively to the B) histological staining in cases where the imaging plane was relatively aligned to the histology cross-section. Hypoxyprobe IHC, a marker of hypoxia, demonstrated darker staining in the tumor regions distant from the CAM membrane (left inset). In comparison, the regions closer to the membrane had reduced hypoxia staining (right inset). Ki-67 staining was prevalent across the depth of the tumor. C) Quantifications of the hypoxyprobe staining (top row) and Ki67 staining (bottom row) for both the HCT-116 and HT-29 cell lines for all treatment groups.
Fig. 6.
Fig. 6.
Quantification of tumor vascularization. A) HCT-116 tumors showed a trend toward reduced blood volume for the cohorts treated with sorafenib, with a significant difference detected in the combination therapy group. B) The intervessel distance for this cell line demonstrated significant decreases for the control group, increased for both chemotherapy treatments, and little change for the sorafenib monotherapy. C) Tumor mean blood velocity trended toward slower flow, in particular for the combination therapy group. D) HT-29 tumors also demonstrated a longitudinal trend of reduced blood volume, but no significance difference was detected. E) Intervessel distance was relatively static for the HT-29 cell line for all treatment groups. F) Mean blood velocity also demonstrated a trend of reduced velocity, but no significance was found.
Fig. 7.
Fig. 7.
Hausdorff box-counting of ULM images. The Hausdorff fractal dimension of ULM vascular maps was estimated using the slope generated using a multi-dimensional box-counting algorithm. Interestingly, all tumor groups treated with the AA agent sorafenib showed a significant increase in Hausdorff dimension. The HT-29 FOLFOX-treated tumors also demonstrated a significant increase in Hausdorff dimension but the opposite trend was noted in HCT-116 tumors treated with FOLFOX.
Fig. 8.
Fig. 8.
Measures of vascular tortuosity. Vascular tortuosity was measured using the distance metric (DM) and the sum of angle metric (SOAM). For the HCT-116 cell line, the A) DM was found to significantly increase in the combination therapy group and B) SOAM measures were found to significantly increase in both sorafenib containing therapies. The HT-29 cell line showed C) a significantly increased DM for FOLFOX treated tumors, and D) increased SOAM in the FOLFOX and combination therapy treated groups.

References

    1. Siegel RL, Miller KD, and Jemal A, “Cancer statistics, 2019,” CA Cancer J. Clin, vol. 69, no. 1, pp. 7–34, 2019, doi: 10.3322/caac.21551.
    1. Van Cutsem E et al. “ESMO consensus guidelines for the management of patients with metastatic colorectal cancer,” Ann. Oncol, vol. 27, no. 8, pp. 1386–1422, 2016, doi: 10.1093/annonc/mdw235.
    1. Saltz LB et al. “Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: A randomized phase III study,” J. Clin. Oncol, vol. 26, no. 12, pp. 2013–2019, Apr. 2008, doi: 10.1200/JCO.2007.14.9930.
    1. Giantonio BJ et al. “Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: Results from the Eastern Cooperative Oncology Group study E3200,” J. Clin. Oncol, vol. 25, no. 12, pp. 1539–1544, Apr. 2007, doi: 10.1200/JCO.2006.09.6305.
    1. Hurwitz H et al. “Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer,” New England J. Med, vol. 350, no. 23, pp. 2335–2342, Jun. 2004, doi: 10.1056/NEJMoa032691.
    1. Kabbinavar F et al. “Phase II, randomized trial comparing bevacizumab plus fluorouracil (FU)/leucovorin (LV) with FU/LV alone in patients with metastatic colorectal cancer,” J. Clin. Oncol, vol. 21, no. 1, pp. 60–65, Jan. 2003, doi: 10.1200/JCO.2003.10.066.
    1. Jain RK, “Normalizing tumor vasculature with anti-angiogenic therapy: A new paradigm for combination therapy,” Nature Med., vol. 7, no. 9, pp. 987–989, Sep. 2001, doi: 10.1038/nm0901-987.
    1. Heldin C-H et al. “High interstitial fluid pressure - An obstacle in cancer therapy,” Nature Rev. Cancer, vol. 4, no. 10, pp. 806–813, Oct. 2004, doi: 10.1038/nrc1456.
    1. Cosse J-P and Michiels C, “Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression,” Anticancer Agents Med. Chem, vol. 8, no. 7, pp. 790–797, Oct. 2008, doi: 10.2174/187152008785914798.
    1. Tabernero J et al. “Sorafenib in combination with oxaliplatin, leucovorin, and fluorouracil (modified FOLFOX6) as first-line treatment of metastatic colorectal cancer: The RESPECT trial,” Clin. Cancer Res, vol. 19, no. 9, pp. 2541–2550, May 2013, doi: 10.1158/1078-0432.CCR-13-0107.
    1. Jain RK et al. “Biomarkers of response and resistance to antiangiogenic therapy,” Nature Rev. Clin. Oncol, vol. 6, no. 6, pp. 327–338, Jun. 2009, doi: 10.1038/nrclinonc.2009.63.
    1. Eisenhauer EA et al. “New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1),” Eur. J. Cancer, vol. 45, no. 2, pp. 228–247, Jan. 2009, doi: 10.1016/j.ejca.2008.10.026.
    1. Miller AB et al. “Reporting results of cancer treatment,” Cancer, vol. 47, no. 1, pp. 207–214, Jan. 1981,doi: 10.1002/1097-0142(19810101)47:1<207::aid-cncr2820470134>;2-6.
    1. Edeline J et al. “Comparison of tumor response by response evaluation criteria in solid tumors (RECIST) and modified RECIST in patients treated with sorafenib for hepatocellular carcinoma,” Cancer, vol. 118, no. 1, pp. 147–156, 2012, doi: 10.1002/cncr.26255.
    1. Tirkes T et al. “Response criteria in oncologic imaging: Review of traditional and new criteria,” Radiographics, vol. 33, no. 5, pp. 1323–1341, Oct. 2013, doi: 10.1148/rg.335125214.
    1. Lencioni R and Llovet JM, “Modified RECIST (mRECIST) assessment for hepatocellular carcinoma,” Semin. Liver Dis, vol. 30, no. 1, pp. 52–60, Feb. 2010, doi: 10.1055/s-0030-1247132.
    1. Choi H et al. “Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: Proposal of new computed tomography response criteria,” J. Clin. Oncol, vol. 25, no. 13, pp. 1753–1759, May 2007, doi: 10.1200/JCO.2006.07.3049.
    1. Lassau N et al. “Validation of dynamic contrast-enhanced ultrasound in predicting outcomes of antiangiogenic therapy for solid tumors: The french multicenter support for innovative and expensive techniques study,” Invest. Radiol, vol. 49, no. 12, pp. 794–800, Dec. 2014, doi: 10.1097/RLI.0000000000000085.
    1. Errico C et al. “Ultrafast ultrasound localization microscopy for deep super-resolution vascular imaging,” Nature, vol. 527, no. 7579, pp. 499–502, Nov. 2015, doi: 10.1038/nature16066.
    1. Christensen-Jeffries K et al. “In vivo acoustic super-resolution and super-resolved velocity mapping using microbubbles,” IEEE Trans. Med. Imag, vol. 34, no. 2, pp. 433–440, Feb. 2015, doi: 10.1109/TMI.2014.2359650.
    1. Couture O et al. “Ultrasound localization microscopy and super-resolution: A state of the art,” IEEE Trans. Ultrasonics, Ferroelect., Freq. Control, vol. 65, no. 8, pp. 1304–1320, Aug. 2018, doi: 10.1109/TUFFC.2018.2850811.
    1. Desailly Y et al. “Resolution limits of ultrafast ultrasound localization microscopy,” Phys. Med. Biol, vol. 60, no. 22, pp.8723–8740, Nov. 2015, doi: 10.1088/0031-9155/60/22/8723.
    1. Ackermann D and Schmitz G, “Detection and tracking of multiple microbubbles in ultrasound B-mode images,” IEEE Trans. Ultrason. Ferroelectr. Freq. Control, vol. 63, no. 1, pp. 72–82, Jan. 2016, doi: 10.1109/TUFFC.2015.2500266.
    1. Opacic T et al. “Motion model ultrasound localization microscopy for preclinical and clinical multiparametric tumor characterization,” Nature Commun., vol. 9, no. 1, 2018, Art. no. 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, vol. 66, no. 3, pp.517–526, Mar. 2019, doi: 10.1109/TUFFC.2018.2872067.
    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, vol. 7, no. 1, pp. 196–204, Jan. 2017, doi: 10.7150/thno.16899.
    1. Bullitt E et al. “Measuring tortuosity of the intracerebral vasculature from MRA images,” IEEE Trans. Med. Imag, vol. 22, no. 9, pp. 1163–1171, Sep. 2003, doi: 10.1109/TMI.2003.816964.
    1. Lowerison MR et al. “Ultrasound localization microscopy of renal tumor xenografts in chicken embryo is correlated to hypoxia,” Sci. Rep, vol. 10, no. 1, pp. 1–13, Feb. 2020, doi: 10.1038/s41598-020-59338-z.
    1. Baish JW and Jain RK, “Fractals and cancer,” Cancer Res., vol. 60, no. 14, pp. 3683–3688, Jul. 2000.
    1. Gazit Y et al. “Fractal characteristics of tumor vascular architecture during tumor growth and regression,” Microcirculation, vol. 4, no. 4, pp. 395–402, Dec. 1997, doi: 10.3109/10739689709146803.
    1. Meakin P, “Diffusion-controlled cluster formation in 2–6-dimensional space,” Phys. Rev. A, vol. 27, no. 3, pp. 1495–1507, Mar. 1983, doi: 10.1103/PhysRevA.27.1495.
    1. Huang C et al. “Noninvasive contrast-free 3D evaluation of tumor angiogenesis with ultrasensitive ultrasound microvessel imaging,” Sci. Rep, vol. 9, no. 1, pp. 4907, Mar. 2019, doi: 10.1038/s41598-019-41373-0.
    1. Sacco JJ et al. “The average body surface area of adult cancer patients in the U.K.: A multicentre retrospective study,” PLoS One, vol. 5, no. 1, Jan. 2010, Art. no. e8933, doi: 10.1371/journal.pone.0008933.
    1. Meijlink B et al. “The preparation of chicken ex ovo embryos and chorioallantoic membrane vessels as in vivo model for contrast-enhanced ultrasound imaging and microbubble-mediated drug delivery studies,” J. Vis. Exp, no. 168, p. e62076, Feb. 2021, doi: 10.3791/62076.
    1. Desailly Y et al. “Contrast enhanced ultrasound by real-time spatiotemporal filtering of ultrafast images,” Phys. Med. Biol, vol. 62, no. 1, pp. 31–42, July 2017, doi: 10.1088/1361-6560/62/1/31.
    1. Huang C et al. “Short acquisition time super-resolution ultrasound microvessel imaging via microbubble separation,” Sci. Rep, vol. 10, no. 1, Apr. 2020, Art. no. 1, doi: 10.1038/s41598-020-62898-9.
    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, vol. 65, no. 2, pp. 149–167, Feb. 2018, doi: 10.1109/TUFFC.2017.2778941.
    1. Song P et al. “Ultrasound small vessel imaging with block-wise adaptive local clutter filtering,” IEEE Trans. Med. Imag, vol. 36, no. 1, pp. 251–262, Jan. 2017, doi: 10.1109/TMI.2016.2605819.
    1. Song P et al. “Noise equalization for ultrafast plane wave microvessel imaging,” IEEE Trans. Ultrason. Ferroelectr. Freq. Control, vol. 64, no. 11, pp. 1776–1781, Nov. 2017, doi: 10.1109/TUFFC.2017.2748387.
    1. Tang S et al. “Kalman filter–based microbubble tracking for robust super-resolution ultrasound microvessel imaging,” IEEE Trans. Ultrasonics, Ferroelect., Freq. Control, vol. 67, no. 9, pp. 1738–1751, Sep. 2020, doi: 10.1109/TUFFC.2020.2984384.
    1. Jaqaman K et al. “Robust single-particle tracking in live-cell time-lapse sequences,” Nature Methods, vol. 5, no. 8, Aug. 2008, Art. no. 8, doi: 10.1038/nmeth.1237.
    1. Hobby JD, “Smooth, easy to compute interpolating splines,” Discrete Comput. Geom, vol. 1, no. 2, pp. 123–140, Jun. 1986, doi: 10.1007/BF02187690.
    1. Shelton SE et al. “Quantification of microvascular tortuosity during tumor evolution utilizing acoustic angiography,” Ultrasound Med. Biol, vol. 41, no. 7, pp. 1896–1904, Jul. 2015, doi: 10.1016/j.ultrasmedbio.2015.02.012.
    1. Nguyen T, “Hausdorff (Box-Counting) fractal dimension with multi-resolution calculation,” 2021. Accessed: Jul. 12,2021. [Online]. Available: .
    1. Pham N-A et al. “Quantitative image analysis of immunohistochemical stains using a CMYK color model,” Diagn. Pathol, vol. 2, p. 8, Feb. 2007, doi: 10.1186/1746-1596-2-8.
    1. McCarty KS et al. “Estrogen receptor analyses. Correlation of bio-chemical and immunohistochemical methods using monoclonal antireceptor antibodies,” Arch. Pathol. Lab. Med, vol. 109, no. 8, pp. 716–721, Aug. 1985.
    1. R. Core Team, R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing, 2019. [Online]. Available:
    1. Wickham H, Ggplot2: Elegant Graphics for Data Analysis. Springer-Verlag: New York, NY, USA, 2016. [Online]. Available:
    1. Roh SA et al. “Characterization of biological responses of colorectal cancer cells to anticancer regimens,” J. Korean Surg. Soc, vol. 83, no. 1, pp. 21–29, Jul. 2012, doi: 10.4174/jkss.2012.83.1.21.
    1. Grossi V et al. “Sorafenib inhibits p38α activity in colorectal cancer cells and synergizes with the DFG-in inhibitor SB202190 to increase apoptotic response,” Cancer Biol. Ther, vol. 13, no. 14, pp. 1471–1481, Dec. 2012, doi: 10.4161/cbt.22254
    1. Ahluwalia A et al. “Aberrant, ectopic expression of VEGF and VEGF receptors 1 and 2 in malignant colonic epithelial cells. Implications for these cells growth via an autocrine mechanism,” Biochem. Biophysical Res. Commun, vol. 437, no. 4, pp. 515–520, Aug. 2013, doi: 10.1016/j.bbrc.2013.06.096.
    1. Yao K et al. “In vitro hypoxia-conditioned colon cancer cell lines derived from HCT116 and HT29 exhibit altered apoptosis susceptibility and a more angiogenic profile in vivo,” Br. J. Cancer, vol. 93, no. 12, pp. 1356–1363, Dec. 2005, doi: 10.1038/sj.bjc.6602864.
    1. Shinagare AB et al. “Liver metastases in the era of molecular targeted therapy: New faces of treatment response,” Amer. J. Roentgenol, vol. 201, no. 1, pp. W15–W28, Jun. 2013, doi: 10.2214/AJR.12.9498.
    1. Marimpietri D et al. “Synergistic inhibition of human neuroblastoma-related angiogenesis by vinblastine and rapamycin,” Oncogene, vol. 24, no. 45, pp. 6785–6795, Oct. 2005, doi: 10.1038/sj.onc.1208829.
    1. Marimpietri D et al. “Combined therapeutic effects of vinblastine and rapamycin on human neuroblastoma growth, apoptosis, and angiogenesis,” Clin. Cancer. Res, vol. 13, no. 13, pp. 3977–3988, Jul. 2007, doi: 10.1158/1078-0432.CCR-06-2757.
    1. Morgenstern DA et al. “Phase I study of vinblastine and sirolimus in pediatric patients with recurrent or refractory solid tumors,” Pediatr. Blood Cancer, vol. 61, no. 1, pp. 128–133, Jan. 2014, doi: 10.1002/pbc.24656.
    1. Huang C et al. “Super-resolution ultrasound localization microscopy based on a high frame-rate clinical ultrasound scanner: An in-human feasibility study,” Phys. Med. Biol, vol. 66, no. 8, p. 08NT01, Apr. 2021, doi: 10.1088/1361-6560/abef45.

Source: PubMed

Подписаться