Intraoperative intravital microscopy permits the study of human tumour vessels

Daniel T Fisher, Jason B Muhitch, Minhyung Kim, Kurt C Doyen, Paul N Bogner, Sharon S Evans, Joseph J Skitzki, Daniel T Fisher, Jason B Muhitch, Minhyung Kim, Kurt C Doyen, Paul N Bogner, Sharon S Evans, Joseph J Skitzki

Abstract

Tumour vessels have been studied extensively as they are critical sites for drug delivery, anti-angiogenic therapies and immunotherapy. As a preclinical tool, intravital microscopy (IVM) allows for in vivo real-time direct observation of vessels at the cellular level. However, to date there are no reports of intravital high-resolution imaging of human tumours in the clinical setting. Here we report the feasibility of IVM examinations of human malignant disease with an emphasis on tumour vasculature as the major site of tumour-host interactions. Consistent with preclinical observations, we show that patient tumour vessels are disorganized, tortuous and ∼50% do not support blood flow. Human tumour vessel diameters are larger than predicted from immunohistochemistry or preclinical IVM, and thereby have lower wall shear stress, which influences delivery of drugs and cellular immunotherapies. Thus, real-time clinical imaging of living human tumours is feasible and allows for detection of characteristics within the tumour microenvironment.

Conflict of interest statement

J.J.S. is an author on a portable IVM platform with a US patent—‘Clinical Intravital Microscope' serial No. 61/988,626 filed by Roswell Park Cancer Institute. K.C.D is an employee of Spectra Services. The remaining authors declare no competing financial interests.

Figures

Figure 1. Design of a portable intraoperative…
Figure 1. Design of a portable intraoperative IVM system and surgical exposure of tumour microvasculature.
(a) Schematic and (b) photograph of the intravital microscopy unit designed as a mobile system for observations in the OR. Critical systems allowing for stable epifluorescence observation of patient tumours in real-time are highlighted as follows: (1) × 10 objective lens (× 100 total including × 10 subjective lens), (2) fluorescent filter cube, (3) fine focus knob, (4) monitor to observe captured movie in real time, (5) cantilevered arm to extend the system over the patient, (6) heavy movement platform to control fine xy motion of the system during observation, (7) solid granite base to reduce vibrations, (8) locking wheels, allowing mobility and stability, (9) integrated computer system to record observations and store data and (10) fluorescent light source. (c) Schematic and (d) representative photographs, detailing the surgical procedure to allow for observation of tumour microvasculature. Briefly, an incision is made in the skin overlying or adjacent to the tumour (red circle) to allow access for direct tumour observation. The epifluorescent light source is activated and digital movie recording commenced to observe the intravenous injection of fluorescein. Following observation the access site is closed and a standard oncologic resection performed.
Figure 2. Detection of blood flow in…
Figure 2. Detection of blood flow in patient tumour microvasculature.
(a) Representative photomicrographs of tumour microvasculature in patients following fluorescein injection. (b,c) Photomicrographs exported during movie analysis of blood flow velocity. (b) Arrow denotes the trailing edge of a bright fluorescein signal that was utilized to determine blood flow velocity in mm s−1. (c) Measurement of tumour diameters and blood vessel flow rate. White arrows indicate vessels that did not support blood flow during observation, black arrows denote functional vessels and white arrowheads show adipocytes. Brackets depict tumour diameter measurements of non-functional (white) and functional (black) vessels with wavy black lines demonstrating direction of blood flow and flow rate. (ac) Bar is 100 μm.
Figure 3. Comparison of tumour vessel diameters…
Figure 3. Comparison of tumour vessel diameters by different methodologies.
(a) Individual measurements of tumour vessel diameters in patients. (b) Tumour vessel diameters quantified by human IVM and IHC of the same tumour tissue and murine melanoma tumours demonstrate consistently larger diameters detected by IVM. Established methods of estimating human tumour vessel diameters by IHC or mouse IVM produce similar measurements that both significantly underestimate diameters measured by human IVM. (c) The wall shear stress in tumour vessels is larger in murine tumours when compared with patient tumours. (d) Wall shear rate is lower in patient tumour in comparison to patient tumours. (ad) *P<0.05l; NS, non significant (unpaired Student's t-test). n=9 for patients; n=10 for mice. Data is mean±s.e.m.
Figure 4. Vessel diameters are similar in…
Figure 4. Vessel diameters are similar in the periphery and core of tumours.
(a) Marking dye was applied following surgical resection to identify the imaged surface of tumour. Measurements of core (>200 μm from surface) and peripheral (<200 μm from surface) tumour blood vessels was performed on formalin-fixed tumour samples stained using haematoxylin and anti-CD31 mAb. Bar is 100 μm. (b) Comparison of tumour vessel diameters per field (minimum 5 fields) in the periphery (P) versus core (C) in humans and mice, NS, non significant (unpaired Student's t-test). n=3 for patients; n=5 for mice. Data is mean±s.e.m.

References

    1. Jain R. K., Munn L. L. & Fukumura D. Dissecting tumour pathophysiology using intravital microscopy. Nat. Rev. Cancer 2, 266–276 (2002).
    1. Fukumura D., Duda D. G., Munn L. L. & Jain R. K. Tumor microvasculature and microenvironment: novel insights through intravital imaging in pre-clinical models. Microcirculation 17, 206–225 (2010).
    1. Barretto R. P. et al.. Time-lapse imaging of disease progression in deep brain areas using fluorescence microendoscopy. Nat. Med. 17, 223–228 (2011).
    1. McDonald D. M. & Choyke P. L. Imaging of angiogenesis: from microscope to clinic. Nat. Med. 9, 713–725 (2003).
    1. Helmlinger G., Yuan F., Dellian M. & Jain R. K. Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat. Med. 3, 177–182 (1997).
    1. Dewhirst M. W. et al.. Quantification of longitudinal tissue pO2 gradients in window chamber tumours: impact on tumour hypoxia. Br. J. Cancer 79, 1717–1722 (1999).
    1. Zijlstra A., Lewis J., Degryse B., Stuhlmann H. & Quigley J. P. The inhibition of tumor cell intravasation and subsequent metastasis via regulation of in vivo tumor cell motility by the tetraspanin CD151. Cancer Cell 13, 221–234 (2008).
    1. Beerling E., Ritsma L., Vrisekoop N., Derksen P. W. & van Rheenen J. Intravital microscopy: new insights into metastasis of tumors. J. Cell Sci. 124, 299–310 (2011).
    1. Mikucki M. E. et al.. Non-redundant requirement for CXCR3 signalling during tumoricidal T-cell trafficking across tumour vascular checkpoints. Nat. Commun. 6, 7458 (2015).
    1. Wu N. Z., Klitzman B., Dodge R. & Dewhirst M. W. Diminished leukocyte-endothelium interaction in tumor microvessels. Cancer Res. 52, 4265–4268 (1992).
    1. Sumen C., Mempel T. R., Mazo I. B. & von Andrian U. H. Intravital microscopy: visualizing immunity in context. Immunity 21, 315–329 (2004).
    1. Fisher D. T. et al.. IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. J. Clin. Invest. 121, 3846–3859 (2011).
    1. Ellenbroek S. I. & van Rheenen J. Imaging hallmarks of cancer in living mice. Nat. Rev. Cancer 14, 406–418 (2014).
    1. Pittet M. J. & Weissleder R. Intravital imaging. Cell 147, 983–991 (2011).
    1. Palmer G. M. et al.. In vivo optical molecular imaging and analysis in mice using dorsal window chamber models applied to hypoxia, vasculature and fluorescent reporters. Nat. Protoc. 6, 1355–1366 (2011).
    1. Turley R. S. et al.. Bevacizumab-induced alterations in vascular permeability and drug delivery: a novel approach to augment regional chemotherapy for in-transit melanoma. Clin. Cancer Res. 18, 3328–3339 (2012).
    1. Bindea G. et al.. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39, 782–795 (2013).
    1. Algire G. An adaptation of the transparent-chamber technique to the mouse. J. Natl Cancer Inst. 4, 1–11 (1943).
    1. Grigorova I. L. et al.. Cortical sinus probing, S1P1-dependent entry and flow-based capture of egressing T cells. Nat. Immunol. 10, 58–65 (2009).
    1. Moussion C. & Girard J. P. Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules. Nature 479, 542–546 (2011).
    1. von Andrian U. H. Intravital microscopy of the peripheral lymph node microcirculation in mice. Microcirculation 3, 287–300 (1996).
    1. Chen Q. et al.. Fever-range thermal stress promotes lymphocyte trafficking across high endothelial venules via an interleukin 6 trans-signaling mechanism. Nat. Immunol. 7, 1299–1308 (2006).
    1. Alford R. et al.. Toxicity of organic fluorophores used in molecular imaging: literature review. Mol. Imaging 8, 341–354 (2009).
    1. Sturm M. B. et al.. Targeted imaging of esophageal neoplasia with a fluorescently labeled peptide: first-in-human results. Sci. Transl. Med. 5, 184ra161 (2013).
    1. Moore G. E. et al.. The clinical use of fluorescein in neurosurgery; the localization of brain tumors. J. Neurosurg. 5, 392–398 (1948).
    1. van Dam G. M. et al.. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-alpha targeting: first in-human results. Nat. Med. 17, 1315–1319 (2011).
    1. Kalogeromitros D. C. et al.. Allergy skin testing in predicting adverse reactions to fluorescein: a prospective clinical study. Acta Ophthalmol. 89, 480–483 (2011).
    1. Dellinger R. P. et al.. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008. Crit. Care Med. 36, 296–327 (2008).
    1. Chang Y. S. et al.. Mosaic blood vessels in tumors: frequency of cancer cells in contact with flowing blood. Proc. Natl Acad. Sci. USA 97, 14608–14613 (2000).
    1. Stylianopoulos T. & Jain R. K. Combining two strategies to improve perfusion and drug delivery in solid tumors. Proc. Natl Acad. Sci. USA 110, 18632–18637 (2013).
    1. Secomb T. W. et al.. Microangiectasias: structural regulators of lymphocyte transmigration. Proc. Natl Acad. Sci. USA 100, 7231–7234 (2003).
    1. Lawrence M. B., Kansas G. S., Kunkel E. J. & Ley K. Threshold levels of fluid shear promote leukocyte adhesion through selectins (CD62L,P,E). J. Cell Biol. 136, 717–727 (1997).
    1. Melder R. J., Munn L. L., Yamada S., Ohkubo C. & Jain R. K. Selectin- and integrin-mediated T-lymphocyte rolling and arrest on TNF-alpha-activated endothelium: augmentation by erythrocytes. Biophys. J. 69, 2131–2138 (1995).
    1. Schwinn D. A., McIntyre R. W. & Reves J. G. Isoflurane-induced vasodilation: role of the alpha-adrenergic nervous system. Anesth. Analg. 71, 451–459 (1990).
    1. Hoeben A. et al.. Vascular endothelial growth factor and angiogenesis. Pharmacol. Rev. 56, 549–580 (2004).
    1. Tozer G. M. et al.. Mechanisms associated with tumor vascular shut-down induced by combretastatin A-4 phosphate: intravital microscopy and measurement of vascular permeability. Cancer. Res. 61, 6413–6422 (2001).
    1. Ntziachristos V. Going deeper than microscopy: the optical imaging frontier in biology. Nat. Methods 7, 603–614 (2010).
    1. Nagy J. A., Chang S. H., Shih S. C., Dvorak A. M. & Dvorak H. F. Heterogeneity of the tumor vasculature. Semin. Thromb. Hemost. 36, 321–331 (2010).
    1. Less J. R., Skalak T. C., Sevick E. M. & Jain R. K. Microvascular architecture in a mammary carcinoma: branching patterns and vessel dimensions. Cancer Res. 51, 265–273 (1991).
    1. Sennino B. et al.. Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov. 2, 270–287 (2012).
    1. Sorensen A. G. et al.. Increased survival of glioblastoma patients who respond to antiangiogenic therapy with elevated blood perfusion. Cancer Res. 72, 402–407 (2012).
    1. Emblem K. E. et al.. Vessel architectural imaging identifies cancer patient responders to anti-angiogenic therapy. Nat. Med. 19, 1178–1183 (2013).
    1. Folberg R. et al.. The prognostic value of tumor blood vessel morphology in primary uveal melanoma. Ophthalmology 100, 1389–1398 (1993).
    1. Makitie T., Summanen P., Tarkkanen A. & Kivela T. Microvascular loops and networks as prognostic indicators in choroidal and ciliary body melanomas. J. Natl Cancer Inst. 91, 359–367 (1999).
    1. Quester R. & Schroder R. The shrinkage of the human brain stem during formalin fixation and embedding in paraffin. J. Neurosci. Methods 75, 81–89 (1997).
    1. McLaughlin R. A. et al.. Imaging of human lymph nodes using optical coherence tomography: potential for staging cancer. Cancer Res. 70, 2579–2584 (2010).
    1. Konerding M. A., Fait E. & Gaumann A. 3D microvascular architecture of pre-cancerous lesions and invasive carcinomas of the colon. Br. J. Cancer 84, 1354–1362 (2001).
    1. Himburg H. A. et al.. Spatial comparison between wall shear stress measures and porcine arterial endothelial permeability. Am. J. Physiol. Heart. Circ. Physiol. 286, H1916–H1922 (2004).
    1. Primeau A. J., Rendon A., Hedley D., Lilge L. & Tannock I. F. The distribution of the anticancer drug Doxorubicin in relation to blood vessels in solid tumors. Clin. Cancer Res. 11, 8782–8788 (2005).
    1. Weidner N. Chapter 14. Measuring intratumoral microvessel density. Methods Enzymol. 444, 305–323 (2008).

Source: PubMed

3
订阅