Preclinical longitudinal imaging of tumor microvascular radiobiological response with functional optical coherence tomography

Valentin Demidov, Azusa Maeda, Mitsuro Sugita, Victoria Madge, Siddharth Sadanand, Costel Flueraru, I Alex Vitkin, Valentin Demidov, Azusa Maeda, Mitsuro Sugita, Victoria Madge, Siddharth Sadanand, Costel Flueraru, I Alex Vitkin

Abstract

Radiation therapy (RT) is widely used for cancer treatment, alone or in combination with other therapies. Recent RT advances have revived interest in delivering higher dose in fewer fractions, which may invoke both cellular and microvascular damage mechanisms. Microvasculature may thus be a potentially sensitive functional biomarker of RT early response, especially for such emerging RT treatments. However it is difficult to measure directly and non-invasively, and its time course, dose dependencies, and overall importance in tumor control are unclear. We use functional optical coherence tomography for quantitative longitudinal in vivo imaging in preclinical models of human tumor xenografts subjected to 10, 20 and 30 Gy doses, furnishing a detailed assessment of vascular remodeling following RT. Immediate (minutes to tens of minutes) and early (days to weeks) RT responses of microvascular supply, as well as tumor volume and fluorescence intensity, were quantified and demonstrated robust and complex temporal dose-dependent behaviors. The findings were compared to theoretical models proposed in the literature.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
(a) NRG mouse with grown BX-PC3 tumor (blue arrow).The back was shaved prior to DSWC installation. (b) Flipped skin on the opposite side of the back showing the grown tumor during DSWC installation procedure. (c) Mouse with DSWC installed. (d) Same mouse as shown in (c) 9 weeks post-DSWC installation and 7.5 weeks post-RT (single-dose 20 Gy). (e) NRG mouse placed in the small-animal irradiator. 225kVp X-rays were incident from the bottom; white-light images showing (f) tumor location within a DSWC covered with EBT-2 Gafchromic film. (g) EBT-2 Gafchromic film color change after 10 Gy single-dose RT. Note that DSWC glass coverslip retaining ring was removed prior to RT to avoid possible dosimetric complications. Scale bar is 3 mm.
Figure 2
Figure 2
(a) Experimental time course. At day -28 tumor cells were injected into the dorsal skin. DSWC was implanted after the tumor volume reached 3–5 mm in diameter. Tumors were irradiated ~10 days after DSWC installation (day 0 labeled with “R”). Right after irradiation tumor vasculature was monitored within 90 minutes (minutes time scale). For five to eight weeks following irradiation, tumor changes were monitored repeatedly with caliper measurements (tumor volume), svOCT imaging (vasculature) and epi-fluorescence microscope imaging (cancer cell fluorescence). Tumor resection for histological staining was performed at selected post-RT stages in several animals to support and validate the in-vivo observations. White light images of the (b) back and (c) front of the tumor within window chamber. (d) svOCT microvasculature map of the area labeled with black dotted rectangle in (c). (e) Tumor cells Ds-Red fluorescence image. Scale bars are 500 μm.
Figure 3
Figure 3
Schematic diagram of the swept-source OCT system setup with quadrature Mach-Zehnder fiber-based interferometer and optical amplification: SOA - semiconductor optical amplifier, PC - polarization controller, A - fiber attenuator, DB - dual balanced photo-detector, DAQ - data acquisition card, C - collimator, MZ Interferometer - Mach-Zehnder interferometer, L - lens, M - mirror, SG - scanning galvo, CR - circulator.
Figure 4
Figure 4
Real-time imaging of tumor microvasculature via svOCT. (a) White light image of window chamber with tumor tissue under the glass coverslip. (b) OCT volumetric image of (a) taken over a 6 × 6 mm2 field of view. (c) 8 sequential same-location B-scans for further inter-frame comparison required for svOCT analysis. (d) The svOCT algorithm for inter-frame comparison of pixel texture. (e)SVzx vascular cross-section obtained with svOCT algorithm. (f) Vascular en-face two-dimensional projection. (g) Depth-encoded vascular en-face two-dimensional projection. (h) Depth-encoded vascular volume of tissue volume shown in (b).
Figure 5
Figure 5
BX-PC3 pancreatic tumor vasculature development within 16 days after subcutaneous injection of cancer cells into dorsal skin. (a) 3 days; (b) 7 days; (c) 10 days; (d) 16 days after injection. (e) Healthy tissue vasculature for comparison. Scale bars are 0.5 mm.
Figure 6
Figure 6
Immediate (minutes - 10s of minutes) tumor microvascular response within 1.5 hours post-RT 10 Gy single-dose. (a) Before irradiation; (b) 30 min after; (c) 45 min after; (d) 60 min after; (e) 90 min after irradiation. Scale bars are 1 mm. The numbers below each panel represent VVD, relative to the pre-radiation value.
Figure 7
Figure 7
Longitudinal OCT imaging of radiation response of the tumor vasculature to single dose of 20 Gy. svOCT images were taken before and multiple times following irradiation. “Day −0” image was taken 1 hour pre-RT and “Day +0” represents 1.5 hours post-RT. Maximum suppression of tumor vasculature is ~ at day 8, followed by vascular re-growth from the vessels outside the tumor. Scale bars = 1 mm.
Figure 8
Figure 8
Tumor development dynamics as reflected via changes in vascular volume density (VVD), tumor volume and fluorescence intensity after 10, 20 and 30 Gy single-dose local irradiation. (a) Tumor microvasculature response. (b) Tumor volume response. (c) Tumor fluorescence response. Quantified values of VVD, volume and fluorescence pre- and post-RT changes of each tumor were normalized to values of these metrics obtained 1 hour before irradiation of the same tumor. Non-irradiated tumors (0 Gy data) continued to grow and were sacrificed at earlier time points for humane reasons (tumors grew too big and data is not shown for the entire course of 7.5 weeks post-RT). (d) Summary graph for VVD, volume and fluorescence response to 20 Gy single-dose irradiation. In (a)–(d), symbols are experimental points, and lines are a guide for the eye. n = 7–15 per group, error bars = mean ± standard deviation. (e) A recently proposed literature model of tumor growth and corresponding changes in microvasculature following 20 Gy single-dose irradiation (after ref.).
Figure 9
Figure 9
Representative images of H&E (top row) and TUNEL (bottom row) staining for tumors irradiated with (a) 0 Gy; (b) 10 Gy; (c) 20 Gy; (d) 30 Gy. Tumors were resected 2 weeks following irradiation. TUNEL positivity for the whole tumor section was 0.3%, 15.8%, 47.6%, and 97.7% for 0, 10, 20, and 30 Gy, respectively. Scale bar = 200 μm.

References

    1. Hall, E. J. Radiobiology for the Radiologist, 6th Edition. (Lippincott Williams and Wilkins, Philadelphia, 2006).
    1. Lo SS, et al. Stereotactic body radiation therapy: A novel treatment modality. Nat. Rev. Clin. Oncol. 2010;7:44–54. doi: 10.1038/nrclinonc.2009.188.
    1. Timmerman RD, Herman J, Cho LC. Emergence of stereotactic body radiation therapy and its impact on current and future clinical practice. J. Clin. Oncol. 2014;32:2847–2854. doi: 10.1200/JCO.2014.55.4675.
    1. Kim DW, et al. Noninvasive assessment of tumor vasculature response to radiation-mediated, vasculature-targeted therapy using quantified power Doppler sonography: implications for improvement of therapy schedules. J. Ultrasound. Med. 2006;25:1507–1517. doi: 10.7863/jum.2006.25.12.1507.
    1. Park HJ, Griffin RJ, Hui S, Levitt SH, Song CW. Radiation-induced vascular damage in tumors: implications of vascular damage in ablative hypofractionated radiotherapy (SBRT and SRS) Radiat. Res. 2012;177:311–327. doi: 10.1667/RR2773.1.
    1. Song CW, et al. Indirect tumor cell death after high-dose hypofractionated irradiation: Implications for stereotactic body radiation therapy and stereotactic radiation surgery. Int. J. Radiat. Oncol. Biol. Phys. 2015;93:166–172. doi: 10.1016/j.ijrobp.2015.05.016.
    1. Garcia-Barros M, et al. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science. 2003;300:1155–1159. doi: 10.1126/science.1082504.
    1. Fuks A, Kolesnick R. Engaging the vascular component of the tumor response. Cancer Cell. 2005;8:89–91. doi: 10.1016/j.ccr.2005.07.014.
    1. Garcia-Barros M, et al. Impact of stromal sensitivity on radiation response of tumors implanted in scid hosts revisited. Cancer Res. 2010;70:8179–8186. doi: 10.1158/0008-5472.CAN-10-1871.
    1. Kocher M, et al. Computer simulation of cytotoxic and vascular effects of radiosurgery in solid and necrotic brain metastases. Radiother. Oncol. 2000;54:149–156. doi: 10.1016/S0167-8140(99)00168-1.
    1. Hobson B, Denekamp J. Endothelial proliferation in tumours and normal tissues: continuous labelling studies. Br. J. Cancer. 1984;49:405–413. doi: 10.1038/bjc.1984.66.
    1. Barker HE, Paget JTE, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat. Rev. Cancer. 2015;15:409–425. doi: 10.1038/nrc3958.
    1. Stancevic B, et al. Adenoviral transduction of human acid sphingomyelinase into neo-angiogenic endothelium radiosensitizes tumor cure. PLoS ONE. 2013;8:e69025. doi: 10.1371/journal.pone.0069025.
    1. Song CW, et al. Is there indirect cell death involved in response of tumor to SRS and SBRT? Int. J. Radiat. Oncol. Biol. Phys. 2014;89:924–925. doi: 10.1016/j.ijrobp.2014.03.043.
    1. Sperduto PW, Song CW, Kirkpatrick J, Glatstein E. A hypothesis on indirect cell death in the radiosurgery era. Int. J. Radiat. Oncol. Biol. Phys. 2015;91:11–13. doi: 10.1016/j.ijrobp.2014.08.355.
    1. Brown JM, Carlson DJ, Brenner DJ. The tumor radiobiology of SRS and SBRT: Are more than the 5Rs involved? Int. J. Radiat. Oncol. Biol. Phys. 2014;88:254–262. doi: 10.1016/j.ijrobp.2013.07.022.
    1. Karam SD, Bhatia S. The radiobiological targets of SBRT: Tumor cells or endothelial cells? Ann. Transl. Med. 2015;19:290.
    1. Kozin SV, Duda DG, Munn LL, Jain RK. Neovascularization after irradiation: what is the source of newly formed vessels in recurring tumors? J. Natl. Cancer Inst. 2012;104:809–905. doi: 10.1093/jnci/djs239.
    1. Popescu DP, et al. Optical coherence tomography: fundamental principles, instrumental designs and biomedical applications. Biophys. Rev. 2011;3:155–169. doi: 10.1007/s12551-011-0054-7.
    1. Mariampillai A, et al. Optimized speckle variance OCT imaging of microvasculature. Opt. Lett. 2010;35:1257–1259. doi: 10.1364/OL.35.001257.
    1. Mariampillai A, et al. Speckle variance detection of microvasculature using swept-source optical coherence tomography. Opt. Lett. 2008;33:1530–1532. doi: 10.1364/OL.33.001530.
    1. Leung, M. K. K. A platform to monitor tumour cellular and vascular response to radiation therapy by optical coherence tomography and fluorescence microscopy in-vivo. MSc thesis, Medical Biophysics, University of Toronto (2010).
    1. Maeda A, DaCosta RS. Optimization of the dorsal skinfold window chamber model and multi-parametric characterization of tumorassociated vasculature. IntraVital. 2014;3:e27935. doi: 10.4161/intv.27935.
    1. Maeda, A. et al. In vivo optical imaging of tumor and microvascular response to ionizing radiation PLoS ONE7, e42133, 1–15 (2012).
    1. Conroy L, DaCosta RS, Vitkin IA. Quantifying microvasculature with speckle variance OCT. Opt. Lett. 2012;37:3180–3182. doi: 10.1364/OL.37.003180.
    1. Pearson T, et al. Non-obese diabeticrecombination activating gene-1 (NOD-Rag1 null) interleukin (IL)-2 receptor common gamma chain (IL2r gamma null) null mice: A radioresistant model for human lymphohaematopoietic engraftment. Clin. Exp. Immunol. 2008;154:270–284. doi: 10.1111/j.1365-2249.2008.03753.x.
    1. Brunner TB, Nestle U, Grosu A-L, Partridge M. SBRT in pancreatic cancer: What is the therapeutic window? Radiother. Oncol. 2015;114:109–116. doi: 10.1016/j.radonc.2014.10.015.
    1. de Geus, S. W. L. et al. Stereotactic body radiotherapy for unresected pancreatic cancer: A nationwide review. Cancer (in press, 10.1002/cncr.30856 (2017).
    1. Rosati LM, Kumar R, Herman JM. Integration of stereotactic body radiation therapy into the multidisciplinary management of pancreatic cancer. Semin. Radiat. Oncol. 2017;27:256–267. doi: 10.1016/j.semradonc.2017.02.005.
    1. Norrby K. Microvascular density in terms of number and length of microvessel segments per unit tissue volume in mammalian angiogenesis. Microvasc. Res. 1998;55:43–53. doi: 10.1006/mvre.1997.2054.
    1. Tao YK, Kennedy KM, Izatt JA. Velocity-resolved 3D retinal microvessel imaging using single-pass flow imaging spectral domain optical coherence tomography. Opt. Express. 2009;17:4177–4188. doi: 10.1364/OE.17.004177.
    1. Reif R, et al. Quantifying optical microangiography images obtained from a spectral domain optical coherence tomography system. J. Biomed. Imaging. 2012;509783:1–11.
    1. Suetsugu A, et al. Non-invasive fluorescent-protein imaging of orthotopic pancreatic-cancer-patient tumorgraft progression in nude mice. Anticancer Res. 2012;32:3063–3068.
    1. Baron VT, Welsh J, Abedinpour P, Borgström P. Intravital microscopy in the mouse dorsal chamber model for the study of solid tumors. Am. J. Cancer Res. 2011;1:674–686.
    1. Clarkson R, et al. Characterization of image quality and image-guidance performance of a preclinical microirradiator. Med. Phys. 2011;38:845–856. doi: 10.1118/1.3533947.
    1. Mao Y, Sherif S, Flueraru C, Chang S. 3 × 3 Mach-Zehnder interferometer with unbalanced differential detection for full-range swept-source optical coherence tomography. Appl. Opt. 2008;47:2004–2010. doi: 10.1364/AO.47.002004.
    1. Mao Y, Flueraru C, Chang S, Popescu D, Sowa M. High-quality tissue imaging using a catheter-based swept-source optical coherence tomography systems with an integrated semiconductor optical amplifier. IEEE Trans. Instrum. Meas. 2011;60:3376–3383. doi: 10.1109/TIM.2011.2126950.
    1. Fitzpatrick, J.M., Sonka, M. Handbook of Medical Imaging, Volume 2. Medical Image Processing and Analysis. (SPIE Press Book, 2000).
    1. Otsu, N. A. threshold selection method from gray-level histograms. IEEE Trans. Syst, Man Cybern. SMC-962 (1979).
    1. Folkman J. Tumor angiogenesis. Adv. Cancer Res. 1985;43:175–203. doi: 10.1016/S0065-230X(08)60946-X.
    1. Jain RK. Delivery of novel therapeutic agents in tumors: physiological barriers and strategies. J. Nat. Cancer Inst. 1989;81:570–576. doi: 10.1093/jnci/81.8.570.
    1. Vaupel P, Kallinowski F, Okunieff P. Blood flow, oxygenation and nutrient supply, and metabolic microenvironment of human tumors: a review. Cancer Res. 1989;49:6449–6465.
    1. Bruberg KJ, Thuen M, Ruud EBM, Rofstad EK. Fluctuations in pO2 in irradiated human melanoma xenografts. Radiat. Res. 2006;165:16–25. doi: 10.1667/RR3491.1.
    1. Kioi M, et al. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J. Clin. Invest. 2010;120:694–705. doi: 10.1172/JCI40283.
    1. Maeda A, et al. In-vivo imaging reveals significant tumor vascular dysfunction and increased tumor hypoxia-inducible factor-1α expression induced by high single-dose irradiation in a pancreatic tumor model. Int. J. Radiat. Oncol. Biol. Phys. 2017;97:184–194. doi: 10.1016/j.ijrobp.2016.09.005.
    1. Mouthon MA, Vereycken-Holler V, Van der Meeren A, Gaugler MH. Irradiation increases the interactions of platelets with the endothelium in vivo: analysis by intravital microscopy. Radiat. Res. 2003;160:593–599. doi: 10.1667/3068.
    1. Wong HH, Song CW, Levitt SH. Early changes in the functional vasculature of Walker carcinoma 256 following irradiation. Radiology. 1973;108:429–434. doi: 10.1148/108.2.429.
    1. Song CW, Sung JH, Clement JJ, Levitt SH. Vascular changes in neuroblastoma of mice following x-irradiation. Cancer Res. 1974;34:2344–2350.
    1. Ng QS, et al. Acute tumor vascular effects following fractionated radiotherapy in human lung cancer: in vivo whole tumor assessment using volumetric perfusion computed tomography. Int. J. Radiat. Oncol. Biol. Phys. 2007;67:417–424. doi: 10.1016/j.ijrobp.2006.10.005.
    1. Fenton B, Lord EM, Paoni SF. Effects of radiation on tumor intravascular oxygenation, vascular configuration, development of hypoxia, and clonogenic survival. Radiat. Res. 2001;155:360–368. doi: 10.1667/0033-7587(2001)155[0360:EOROTI];2.
    1. Duffy JP, Eibl G, Reber HA, Hines OJ. Influence of hypoxia and neoangiogenesis on the growth of pancreatic cancer. Mol. Cancer. 2003;2:1–10. doi: 10.1186/1476-4598-2-12.
    1. Khan, F. M., Gibbons, J. P. Khan’s the Physics of Radiation Therapy, 5thEdition. (Wolters Kluwer, 2014).
    1. Jhingran A, et al. Tracing conidial fate and measuring host cell antifungal activity using a reporter of microbial viability in the lung. Cell Reports. 2012;2:1762–1773. doi: 10.1016/j.celrep.2012.10.026.
    1. Denekamp J. Vascular endothelium as the vulnerable element in tumours. Acta Radiol. Oncol. 1984;23:217–225. doi: 10.3109/02841868409136015.
    1. Kumar V, et al. Radiomics: the process and the challenges. Magn. Reson. Imaging. 2012;30:1234–1248. doi: 10.1016/j.mri.2012.06.010.
    1. Kang J, Schwartz R, Flickinger J, Beriwal S. Machine learning approaches for predicting radiation therapy outcomes: A Clinician’sPerspective. Int. J. Radiation. Oncol. Biol. Phys. 2015;93:1127–1135. doi: 10.1016/j.ijrobp.2015.07.2286.
    1. Davoudi B, et al. Quantitative assessment of oral microstructural and microvascular changes in late oral radiation toxicity, using noninvasive in-vivo optical coherence tomography. Photon. Lasers Med. 2015;5:21–32.
    1. Maslennikova, A. V. et al. In-vivo patient study of microvascular changes in irradiated oral mucosa using optical coherence tomography. Int. J. Radiation. Oncol. Biol. Phys (in press).

Source: PubMed

Подписаться