Real-time near-infrared fluorescence imaging using cRGD-ZW800-1 for intraoperative visualization of multiple cancer types

Henricus J M Handgraaf, Martin C Boonstra, Hendrica A J M Prevoo, Joeri Kuil, Mark W Bordo, Leonora S F Boogerd, Babs G Sibinga Mulder, Cornelis F M Sier, Maaike L Vinkenburg-van Slooten, A Rob P M Valentijn, Jacobus Burggraaf, Cornelis J H van de Velde, John V Frangioni, Alexander L Vahrmeijer, Henricus J M Handgraaf, Martin C Boonstra, Hendrica A J M Prevoo, Joeri Kuil, Mark W Bordo, Leonora S F Boogerd, Babs G Sibinga Mulder, Cornelis F M Sier, Maaike L Vinkenburg-van Slooten, A Rob P M Valentijn, Jacobus Burggraaf, Cornelis J H van de Velde, John V Frangioni, Alexander L Vahrmeijer

Abstract

Incomplete resections and damage to critical structures increase morbidity and mortality of patients with cancer. Targeted intraoperative fluorescence imaging aids surgeons by providing real-time visualization of tumors and vital structures. This study evaluated the tumor-targeted zwitterionic near-infrared fluorescent peptide cRGD-ZW800-1 as tracer for intraoperative imaging of multiple cancer types. cRGD-ZW800-1 was validated in vitro on glioblastoma (U-87 MG) and colorectal (HT-29) cell lines. Subsequently, the tracer was tested in orthotopic mouse models with HT-29, breast (MCF-7), pancreatic (BxPC-3), and oral (OSC-19) tumors. Dose-ranging studies, including doses of 0.25, 1.0, 10, and 30 nmol, in xenograft tumor models suggest an optimal dose of 10 nmol, corresponding to a human equivalent dose of 63 μg/kg, and an optimal imaging window between 2 and 24 h post-injection. The mean half-life of cRGD-ZW800-1 in blood was 25 min. Biodistribution at 4 h showed the highest fluorescence signals in tumors and kidneys. In vitro and in vivo competition experiments showed significantly lower fluorescence signals when U-87 MG cells (minus 36%, p = 0.02) or HT-29 tumor bearing mice (TBR at 4 h 3.2 ± 0.5 vs 1.8 ± 0.4, p = 0.03) were simultaneously treated with unlabeled cRGD. cRGD-ZW800-1 visualized in vivo all colorectal, breast, pancreatic, and oral tumor xenografts in mice. Screening for off-target interactions, cRGD-ZW800-1 showed only inhibition of COX-2, likely due to binding of cRGD-ZW800-1 to integrin αVβ3. Due to its recognition of various integrins, which are expressed on malignant and neoangiogenic cells, it is expected that cRGD-ZW800-1 will provide a sensitive and generic tool to visualize cancer during surgery.

Keywords: RGD; fluorescence-guided surgery; in vivo diagnosis; integrins; preclinical validation.

Conflict of interest statement

CONFLICTS OF INTEREST

John V. Frangioni is currently CEO of Curadel, Curadel ResVet Imaging, and Curadel Surgical Innovations, which are for-profit companies that have licensed FLARE ® technology from Beth Israel Deaconess Medical Center. All other authors have no conflicts of interests or financial ties to disclose.

Figures

Figure 1. Binding assay and in vitro…
Figure 1. Binding assay and in vitro and in vivo competition experiments
(A) Binding assay of cRGD-ZW800-1 on intermediate integrin-expressing HT-29 cells shows an almost linear increase in fluorescence intensity with increasing applied concentrations. In addition, fluorescence signals are increased when cells are incubated at 37°C compared to 4°C with the various concentrations (p < 0.0001). This may be the result of enhanced internalization of the agent. (B) In vitro competition experiment (1:1 ratio) using unlabeled cRGD (competition group) showing decreased NIR fluorescent signals in both the αvβ3 positive U-87 MG (*p < 0.05) as well as the αvβ3 negative HT-29 cell line compared to the control without unlabeled cRGD. Healthy colon was used as background. (C) In vivo competition experiment using a 200 times higher dose of unlabeled cRGD compared to the dose of cRGD-ZW800-1 (1.24 mg, 2.0 μmol vs. 15.5 μg, 10 nmol) in the orthotopic HT-29 model. Compared to mice in the control group, a significant decrease in TBR was seen at 4 h (*p = 0.02) and 24 h (** p = 0.007). Healthy colon was used as background.
Figure 2. Near-infrared fluorescence imaging of colorectal…
Figure 2. Near-infrared fluorescence imaging of colorectal cancer (HT-29)
(A) Representative images captured using the original FLARE® prototype of the various concentrations at 4 h post injection. For the 30 nmol dose group the exposure time had to be set at 50 msec due to saturation of the near-infrared fluorescence images. For the other dose groups 200 msec was sufficient. NIR fluorescence signals are clearly visible in 10 and 30 nmol groups only. Arrow = tumor; arrowhead = bladder. (B) Near-infrared fluorescence imaging at 4 h post injection showed sufficient tumor-to-background ratios (i.e. ≥ 2) already in the 0.25 nmol (0.39 μg) dose group using the Pearl®. Surrounding tissue was used as background. (C) Near-infrared fluorescence imaging using the Pearl® at 24 h post injection still showed sufficient tumor-to-background ratios in the 1.0, 10 and 30 nmol dose groups. Healthy tongue tissue was used as background.
Figure 3. Near-infrared fluorescence imaging of pancreatic…
Figure 3. Near-infrared fluorescence imaging of pancreatic (BxPC-3) and oral cancer (OSC-19)
(A) Upper panel: example showing the in vivo images of 30 nmol cRGD-ZW800-1 in the orthotopic pancreas model at 4 h post injection using the original FLARE® prototype. Arrow = tumor. Lower panel: NIR fluorescence microscopy of the tumor shows fluorescence inside the cells. (B) Mean tumor-to-background ratios at 4 h post injection in the BXPC-3 model with the Pearl®. Skin was used as background. (C) Upper panel: example of images from the orthotopic OSC-19 model at 4 h using the original FLARE® prototype. Arrow = tumor. Lower panel: NIR fluorescence microscopy shows the border of a resected OSC-19 tumor. Clear fluorescent demarcation between normal and tumor tissue is shown (white dotted line). T = tumor; N = normal tissue surrounding the tumor. (D) Mean tumor-to-background ratios of the optimal dose 10 nmol at 4 h in the orthotopic head-and-neck cancer model using the Pearl®.
Figure 4. Near-infrared fluorescence imaging of breast…
Figure 4. Near-infrared fluorescence imaging of breast cancer (MCF-7)
(A) Representative images captured with the Pearl® of the various time points after administration of 10 nmol cRGD-ZW800-1 in a xenograft breast cancer model. Arrowhead: bladder, arrows: tumors. All images are identically scaled. (B) Tumor-to-background ratios over time measured using the Pearl®. (C) Fluorescence intensity of tumors compared with 0.5 h post injection using the Pearl®.
Figure 5. Biodistribution of cRGD-ZW800-1
Figure 5. Biodistribution of cRGD-ZW800-1
(A) In vivo fluorescence intensity of BxPC-3 and HT-29 tumors at 4 h post injection of several doses of cRGD-ZW800-1 using the Pearl®. Significant differences (*p < 0.05; **p < 0.01) were seen in the 10 nmol dose group (p < 0.05). (B) The ex vivo fluorescence intensity of BxPC-3 tumors were used to calculate the organ-to-tumor ratios at 4 h post injection. Compared to the 10 nmol dose group, significant differences were seen between kidneys (1.0 nmol: p < 0.05; 30 nmol: p < 0.01 ), liver (1.0 nmol: p < 0.05) and large intestines (1.0 nmol: p < 0.05). In all dose groups, tumors were more than twice as fluorescent compared to skin, intestines, lungs, muscle, brain, gallbladder, and blood. (C) In vivo fluorescence intensity of HT-29 tumors at 4 and 24 h post injection of several doses. Significant differences (*) were seen in the 10 nmol dose group (p < 0.05). (D) The ex vivo fluorescence intensity of HT-29 tumors were used to calculate the organ-to-tumor ratios at 24 h post injection. Compared to the 10 nmol dose group, significant differences (*) were seen between kidneys (1.0 nmol and 30 nmol: p < 0.05). In all dose groups, tumors were more than twice as fluorescent compared to lungs, pancreas, muscle, gallbladder, brain, and blood.
Figure 6. Pharmacokinetics of 10 nmol cRGD-ZW800-1
Figure 6. Pharmacokinetics of 10 nmol cRGD-ZW800-1
Graph shows the mean absolute concentrations (in μM) after intravenous administration of 10 nmol cRGD-ZW800-1 up to 240 min post injection. 1 AUC calculated via trapezoid rule. 2 Half-life calculated via ln(2)/k10.

References

    1. Vahrmeijer AL, Hutteman M, van der Vorst JR, van de Velde CJ, Frangioni JV. Image-guided cancer surgery using near-infrared fluorescence. Nat Rev Clin Oncol. 2013;10:507–518.
    1. Sevick-Muraca EM, Houston JP, Gurfinkel M. Fluorescence-enhanced, near infrared diagnostic imaging with contrast agents. Curr Opin Chem Biol. 2002;6:642–650.
    1. Ishizawa T, Fukushima N, Shibahara J, Masuda K, Tamura S, Aoki T, Hasegawa K, Beck Y, Fukayama M, Kokudo N. Real-time identification of liver cancers by using indocyanine green fluorescent imaging. Cancer. 2009;115:2491–2504.
    1. Rosenthal EL, Warram JM, de Boer E, Chung TK, Korb ML, Brandwein-Gensler M, Strong TV, Schmalbach CE, Morlandt AB, Agarwal G, Hartman YE, Carroll WR, Richman JS, et al. Safety and Tumor Specificity of Cetuximab-IRDye800 for Surgical Navigation in Head and Neck Cancer. Clin Cancer Res. 2015;21:3658–3666.
    1. Warram JM, de Boer E, Sorace AG, Chung TK, Kim H, Pleijhuis RG, van Dam GM, Rosenthal EL. Antibody-based imaging strategies for cancer. Cancer Metastasis Rev. 2014;33:809–822.
    1. Frangioni JV. New technologies for human cancer imaging. J Clin Oncol. 2008;26:4012–4021.
    1. Burggraaf J, Kamerling IM, Gordon PB, Schrier L, de Kam ML, Kales AJ, Bendiksen R, Indrevoll B, Bjerke RM, Moestue SA, Yazdanfar S, Langers AM, Swaerd-Nordmo M, et al. Detection of colorectal polyps in humans using an intravenously administered fluorescent peptide targeted against c-Met. Nat Med. 2015;21:955–961.
    1. Hoogstins CE, Tummers QR, Gaarenstroom KN, de Kroon CD, Trimbos JB, Bosse T, Smit VT, Vuyk J, van de Velde CJ, Cohen AF, Low PS, Burggraaf J, Vahrmeijer AL. A Novel Tumor-Specific Agent for Intraoperative Near-Infrared Fluorescence Imaging: A Translational Study in Healthy Volunteers and Patients with Ovarian Cancer. Clin Cancer Res. 2016;22:2929–2938.
    1. Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10:9–22.
    1. Schittenhelm J, Klein A, Tatagiba MS, Meyermann R, Fend F, Goodman SL, Sipos B. Comparing the expression of integrins alphavbeta3, alphavbeta5, alphavbeta6, alphavbeta8, fibronectin and fibrinogen in human brain metastases and their corresponding primary tumors. Int J Clin Exp Pathol. 2013;6:2719–2732.
    1. Sharma R, Kallur KG, Ryu JS, Parameswaran RV, Lindman H, Avril N, Gleeson FV, Lee JD, Lee KH, O'Doherty MJ, Groves AM, Miller MP, Somer EJ, et al. Multicenter Reproducibility of 18F-Fluciclatide PET Imaging in Subjects with Solid Tumors. J Nucl Med. 2015;56:1855–1861.
    1. Beer AJ, Haubner R, Sarbia M, Goebel M, Luderschmidt S, Grosu AL, Schnell O, Niemeyer M, Kessler H, Wester HJ, Weber WA, Schwaiger M. Positron emission tomography using [18F]Galacto-RGD identifies the level of integrin alpha(v)beta3 expression in man. Clin Cancer Res. 2006;12:3942–3949.
    1. Beer AJ, Lorenzen S, Metz S, Herrmann K, Watzlowik P, Wester HJ, Peschel C, Lordick F, Schwaiger M. Comparison of integrin alphaVbeta3 expression and glucose metabolism in primary and metastatic lesions in cancer patients: a PET study using 18F-galacto-RGD and 18F-FDG. J Nucl Med. 2008;49:22–29.
    1. Beer AJ, Niemeyer M, Carlsen J, Sarbia M, Nahrig J, Watzlowik P, Wester HJ, Harbeck N, Schwaiger M. Patterns of alphavbeta3 expression in primary and metastatic human breast cancer as shown by 18F-Galacto-RGD PET. J Nucl Med. 2008;49:255–259.
    1. Verbeek FP, van der Vorst JR, Tummers QR, Boonstra MC, de Rooij KE, Lowik CW, Valentijn AR, van de Velde CJ, Choi HS, Frangioni JV, Vahrmeijer AL. Near-infrared fluorescence imaging of both colorectal cancer and ureters using a low-dose integrin targeted probe. Ann Surg Oncol. 2014;21(Suppl 4):S528–537.
    1. Choi HS, Gibbs SL, Lee JH, Kim SH, Ashitate Y, Liu F, Hyun H, Park G, Xie Y, Bae S, Henary M, Frangioni JV. Targeted zwitterionic near-infrared fluorophores for improved optical imaging. Nat Biotechnol. 2013;31:148–153.
    1. Ruoslahti E. RGD and other recognition sequences for integrins. Annu Rev Cell Dev Biol. 1996;12:697–715.
    1. Brooks PC, Montgomery AM, Rosenfeld M, Reisfeld RA, Hu T, Klier G, Cheresh DA. Integrin alpha v beta 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell. 1994;79:1157–1164.
    1. Humphries JD, Byron A, Humphries MJ. Integrin ligands at a glance. J Cell Sci. 2006;119:3901–3903.
    1. Fang J, Nakamura H, Maeda H. The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Adv Drug Deliv Rev. 2011;63:136–151.
    1. Huang R, Vider J, Kovar JL, Olive DM, Mellinghoff IK, Mayer-Kuckuk P, Kircher MF, Blasberg RG. Integrin alphavbeta3-targeted IRDye 800CW near-infrared imaging of glioblastoma. Clin Cancer Res. 2012;18:5731–5740.
    1. Ke S, Zhang F, Wang W, Qiu X, Lin J, Cameron AG, Zou C, Gao X, Zou C, Zhu VF, Li M. Multiple target-specific molecular imaging agents detect liver cancer in a preclinical model. Curr Mol Med. 2012;12:944–951.
    1. Yoon Y, Mohs AM, Mancini MC, Nie S, Shim H. Combination of an Integrin-Targeting NIR Tracer and an Ultrasensitive Spectroscopic Device for Intraoperative Detection of Head and Neck Tumor Margins and Metastatic Lymph Nodes. Tomography. 2016;2:215–222.
    1. Cao J, Wan S, Tian J, Li S, Deng D, Qian Z, Gu Y. Fast clearing RGD-based near-infrared fluorescent probes for in vivo tumor diagnosis. Contrast Media Mol Imaging. 2012;7:390–402.
    1. Cheng H, Chi C, Shang W, Rengaowa S, Cui J, Ye J, Jiang S, Mao Y, Zeng C, Huo H, Chen L, Tian J. Precise integrin-targeting near-infrared imaging-guided surgical method increases surgical qualification of peritoneal carcinomatosis from gastric cancer in mice. Oncotarget. 2017;8:6258–6272. doi: 10.18632/oncotarget.14058.
    1. Choi HS, Nasr K, Alyabyev S, Feith D, Lee JH, Kim SH, Ashitate Y, Hyun H, Patonay G, Strekowski L, Henary M, Frangioni JV. Synthesis and in vivo fate of zwitterionic near-infrared fluorophores. Angew Chem Int Ed Engl. 2011;50:6258–6263.
    1. Sasaki Y, Iwama R, Sato T, Heishima K, Shimamura S, Ichijo T, Satoh H, Furuhama K. Estimation of glomerular filtration rate in conscious mice using a simplified equation. Physiol Rep. 2014:2.
    1. Morgan MR, Hamidi H, Bass MD, Warwood S, Ballestrem C, Humphries MJ. Syndecan-4 phosphorylation is a control point for integrin recycling. Dev Cell. 2013;24:472–485.
    1. Hyun H, Owens EA, Narayana L, Wada H, Gravier J, Bao K, Frangioni JV, Choi HS, Henary M. Central C-C Bonding Increases Optical and Chemical Stability of NIR Fluorophores. RSC Adv. 2014;4:58762–58768.
    1. Ruegg C, Dormond O, Mariotti A. Endothelial cell integrins and COX-2: mediators and therapeutic targets of tumor angiogenesis. Biochim Biophys Acta. 2004;1654:51–67.
    1. Crofford LJ, Breyer MD, Strand CV, Rushitzka F, Brune K, Farkouh ME, Simon LS. Cardiovascular effects of selective COX-2 inhibition: is there a class effect? The International COX-2 Study Group. J Rheumatol. 2006;33:1403–1408.
    1. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J. 2008;22:659–661.
    1. FDA Estimating the safe starting dose in clinical trials for therapeutics in adult healthy volunteers. pp. 2340–2341.
    1. Piha-Gossack A, Sossin W, Reinhardt DP. The evolution of extracellular fibrillins and their functional domains. PLoS One. 2012;7:e33560.
    1. Beer AJ, Haubner R, Goebel M, Luderschmidt S, Spilker ME, Wester HJ, Weber WA, Schwaiger M. Biodistribution and pharmacokinetics of the alphavbeta3-selective tracer 18F-galacto-RGD in cancer patients. J Nucl Med. 2005;46:1333–1341.
    1. van Driel PB, van de Giessen M, Boonstra MC, Snoeks TJ, Keereweer S, Oliveira S, van de Velde CJ, Lelieveldt BP, Vahrmeijer AL, Lowik CW, Dijkstra J. Characterization and evaluation of the artemis camera for fluorescence-guided cancer surgery. Mol Imaging Biol. 2015;17:413–423.
    1. Stupp R, Hegi ME, Gorlia T, Erridge SC, Perry J, Hong YK, Aldape KD, Lhermitte B, Pietsch T, Grujicic D, Steinbach JP, Wick W, Tarnawski R, et al. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071–22072 study): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15:1100–1108.
    1. Boonstra MC, Tolner B, Schaafsma BE, Boogerd LS, Prevoo HA, Bhavsar G, Kuppen PJ, Sier CF, Bonsing BA, Frangioni JV, van de Velde CJ, Chester KA, Vahrmeijer AL. Preclinical evaluation of a novel CEA-targeting near-infrared fluorescent tracer delineating colorectal and pancreatic tumors. Int J Cancer. 2015;137:1910–1920.
    1. van Driel PB, van der Vorst JR, Verbeek FP, Oliveira S, Snoeks TJ, Keereweer S, Chan B, Boonstra MC, Frangioni JV, van Bergen en Henegouwen PM, Vahrmeijer AL, Lowik CW. Intraoperative fluorescence delineation of head and neck cancer with a fluorescent anti-epidermal growth factor receptor nanobody. Int J Cancer. 2014;134:2663–2673.
    1. Bowes J, Brown AJ, Hamon J, Jarolimek W, Sridhar A, Waldron G, Whitebread S. Reducing safety-related drug attrition: the use of in vitro pharmacological profiling. Nat Rev Drug Discov. 2012;11:909–922.
    1. Tseng W, Leong X, Engleman E. Orthotopic mouse model of colorectal cancer. J Vis Exp. 2007:484.
    1. Kim MP, Evans DB, Wang H, Abbruzzese JL, Fleming JB, Gallick GE. Generation of orthotopic and heterotopic human pancreatic cancer xenografts in immunodeficient mice. Nat Protoc. 2009;4:1670–1680.

Source: PubMed

3
Abonneren