First-in-human intraoperative near-infrared fluorescence imaging of glioblastoma using cetuximab-IRDye800

Sarah E Miller, Willemieke S Tummers, Nutte Teraphongphom, Nynke S van den Berg, Alifia Hasan, Robert D Ertsey, Seema Nagpal, Lawrence D Recht, Edward D Plowey, Hannes Vogel, Griffith R Harsh, Gerald A Grant, Gordon H Li, Eben L Rosenthal, Sarah E Miller, Willemieke S Tummers, Nutte Teraphongphom, Nynke S van den Berg, Alifia Hasan, Robert D Ertsey, Seema Nagpal, Lawrence D Recht, Edward D Plowey, Hannes Vogel, Griffith R Harsh, Gerald A Grant, Gordon H Li, Eben L Rosenthal

Abstract

Introduction: Maximizing extent of surgical resection with the least morbidity remains critical for survival in glioblastoma patients, and we hypothesize that it can be improved by enhancements in intraoperative tumor detection. In a clinical study, we determined if therapeutic antibodies could be repurposed for intraoperative imaging during resection.

Methods: Fluorescently labeled cetuximab-IRDye800 was systemically administered to three patients 2 days prior to surgery. Near-infrared fluorescence imaging of tumor and histologically negative peri-tumoral tissue was performed intraoperatively and ex vivo. Fluorescence was measured as mean fluorescence intensity (MFI), and tumor-to-background ratios (TBRs) were calculated by comparing MFIs of tumor and histologically uninvolved tissue.

Results: The mean TBR was significantly higher in tumor tissue of contrast-enhancing (CE) tumors on preoperative imaging (4.0 ± 0.5) compared to non-CE tumors (1.2 ± 0.3; p = 0.02). The TBR was higher at a 100 mg dose than at 50 mg (4.3 vs. 3.6). The smallest detectable tumor volume in a closed-field setting was 70 mg with 50 mg of dye and 10 mg with 100 mg. On sections of paraffin embedded tissues, fluorescence positively correlated with histological evidence of tumor. Sensitivity and specificity of tumor fluorescence for viable tumor detection was calculated and fluorescence was found to be highly sensitive (73.0% for 50 mg dose, 98.2% for 100 mg dose) and specific (66.3% for 50 mg dose, 69.8% for 100 mg dose) for viable tumor tissue in CE tumors while normal peri-tumoral tissue showed minimal fluorescence.

Conclusion: This first-in-human study demonstrates the feasibility and safety of antibody based imaging for CE glioblastomas.

Keywords: Antibody-based imaging; Brain neoplasms; Cetuximab; Fluorescence; Glioblastoma; Image-guided surgery; Phase 1.

Conflict of interest statement

The authors have no relevant conflicts of interest to disclose.

Figures

Fig. 1
Fig. 1
Trial imaging workflow. Real-time fluorescent imaging was performed in the operating room (1) using a wide-field NIR imaging device on day 2 following cetuximab-IRDye800 infusion. 2 A closed-field NIR imaging system was used to image the resected specimens ex-vivo. 3 Specimens were sectioned and scanned in surgical pathology using a fluorescence scanning system to correlated fluorescence with histological tissue classification
Fig. 2
Fig. 2
Representative intraoperative fluorescent images and associated pre-operative Magnetic Resonance Images (MRIs). Fluorescence image following tumor exposure in a patient 1, b patient 2, and c patient 3 and associated MRIs below
Fig. 3
Fig. 3
Fluorescent images of serially cut fresh tumor and histologically normal tissue imaged in the closed-field setting. Serial sections from a patient 1 and b patient 2. Minimal detectable tumor weights were found to be 70 and 10 mg for patient 1 (low dose) and patient 2 (high dose), respectively
Fig. 4
Fig. 4
MFI of tumor tissue compared to histologically normal and necrotic tumor tissue in glioblastoma patients. a MFI for patient 1 and patient 2 combined, b MFI of patient 1 only (low dose patient), c MFI of patient 2 only (high dose patient) (*P < 0.01, **P < 0.001), d TBR of patient 1 and 2 combined, patient 1 alone, patient 2 alone
Fig. 5
Fig. 5
Representative images and statistical analysis of histological correlation with fluorescence. H&E stain, EGFR and fluorescent images of representative sections containing histologically normal tissue (a) and sections containing tumor and necrotic tissue (b). ROC curves for low (c) and high (d) dose CE glioblastoma patients and associated sensitivity and specificity calculations

References

    1. Thakkar JP, Dolecek TA, Horbinski C, et al. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol Biomark Prev. 2014;23(10):1985–1996. doi: 10.1158/1055-9965.EPI-14-0275.
    1. Lacroix M, Abi-Said D, Fourney DR, et al. A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. J Neurosurg. 2001;95(2):190–198. doi: 10.3171/jns.2001.95.2.0190.
    1. Grabowski MM, Recinos PF, Nowacki AS, et al. Residual tumor volume versus extent of resection: predictors of survival after surgery for glioblastoma. J Neurosurg. 2014;121(5):1115–1123. doi: 10.3171/2014.7.JNS132449.
    1. Nimsky C, Ganslandt O, Cerny S, Hastreiter P, Greiner G, Fahlbusch R (2000) Quantification of, visualization of, and compensation for brain shift using intraoperative magnetic resonance imaging. Neurosurgery 47(5):1070–1079. . Accessed 16 Oct 2017
    1. Cortnum S, Laursen RJ (2012) Fluorescence-guided resection of gliomas. Dan Med J 59(8):A4460. . Accessed 16 Oct 2017
    1. Tykocki T, Michalik R, Bonicki W, Nauman P (2012) Fluorescence-guided resection of primary and recurrent malignant gliomas with 5-aminolevulinic acid. Preliminary results. Neurol Neurochir Pol 46(1):47–51. . Accessed 16 Oct 2017
    1. Stummer W, Pichlmeier U, Meinel T, et al. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006;7(5):392–401. doi: 10.1016/S1470-2045(06)70665-9.
    1. Vahrmeijer AL, Hutteman M, van der Vorst JR, van de Velde CJH, Frangioni JV. Image-guided cancer surgery using near-infrared fluorescence. Nat Rev Clin Oncol. 2013;10(9):507–518. doi: 10.1038/nrclinonc.2013.123.
    1. Rosenthal EL, Warram JM, de Boer E, et al. Safety and tumor specificity of cetuximab-IRDye800 for surgical navigation in head and neck cancer. Clin Cancer Res. 2015;21(16):3658–3666. doi: 10.1158/1078-0432.CCR-14-3284.
    1. Lamberts LE, Koch M, de Jong JS, et al. Tumor-specific uptake of fluorescent bevacizumab–IRDye800CW microdosing in patients with primary breast cancer: a phase i feasibility study. Clin Cancer Res. 2017;23(11):2730–2741. doi: 10.1158/1078-0432.CCR-16-0437.
    1. Warram JM, de Boer E, Korb M, et al. Fluorescence-guided resection of experimental malignant glioma using cetuximab-IRDye 800CW. Br J Neurosurg. 2015;29(6):850–858. doi: 10.3109/02688697.2015.1056090.
    1. Heimberger AB, Hlatky R, Suki D, et al. Prognostic effect of epidermal growth factor receptor and egfrviii in glioblastoma multiforme patients. Clin Cancer Res. 2005;11(4):1462–1466. doi: 10.1158/1078-0432.CCR-04-1737.
    1. Bredel M, Pollack IF, Hamilton RL, James CD (1999) Epidermal growth factor receptor expression and gene amplification in high-grade non-brainstem gliomas of childhood. Clin Cancer Res 5(7):1786–1792. . Accessed 16 Oct 2017
    1. Moore LS, Rosenthal EL, de Boer E, et al. Effects of an unlabeled loading dose on tumor-specific uptake of a fluorescently labeled antibody for optical surgical navigation. Mol Imaging Biol. 2017;19(4):610–616. doi: 10.1007/s11307-016-1022-1.
    1. Zinn KR, Korb M, Samuel S, et al. IND-Directed safety and biodistribution study of intravenously injected cetuximab-IRDye800 in cynomolgus macaques. Mol Imaging Biol. 2015;17(1):49–57. doi: 10.1007/s11307-014-0773-9.
    1. Rosenthal EL, Moore LS, Tipirneni K, et al. Sensitivity and specificity of cetuximab-IRDye800CW to identify regional metastatic disease in head and neck cancer. Clin Cancer Res. 2017;23(16):4744–4752. doi: 10.1158/1078-0432.CCR-16-2968.
    1. Tummers W, Miller S, Teraphongphom N et al (2018) Intraoperative pancreatic cancer detection using tumor-specific multimodality molecular imaging. Ann Surg Oncol (In press)
    1. Scott JN, Brasher PMA, Sevick RJ, Rewcastle NB, Forsyth PA (2002) How often are nonenhancing supratentorial gliomas malignant? A population study. Neurology 59(6):947–949. . Accessed 16 Oct 2017
    1. Hirschberg H, Samset E, Hol PK, Tillung T, Lote K. Impact of intraoperative MRI on the surgical results for high-grade gliomas. Minim Invasive Neurosurg. 2005;48(2):77–84. doi: 10.1055/s-2004-830225.
    1. Shah MN, Leonard JR, Inder G, et al. Intraoperative magnetic resonance imaging to reduce the rate of early reoperation for lesion resection in pediatric neurosurgery. J Neurosurg Pediatr. 2012;9(3):259–264. doi: 10.3171/2011.12.PEDS11227.
    1. Senft C, Bink A, Franz K, Vatter H, Gasser T, Seifert V. Intraoperative MRI guidance and extent of resection in glioma surgery: a randomised, controlled trial. Lancet Oncol. 2011;12(11):997–1003. doi: 10.1016/S1470-2045(11)70196-6.
    1. Minamikawa T, Matsuo H, Kato Y, et al. Simplified and optimized multispectral imaging for 5-ALA-based fluorescence diagnosis of malignant lesions. Sci Rep. 2016;6(1):25530. doi: 10.1038/srep25530.
    1. Miyatake S-I, Kuroiwa T, Kajimoto Y, Miyashita M, Tanaka H, Tsuji M. Fluorescence of non-neoplastic, magnetic resonance imaging-enhancing tissue by 5-aminolevulinic acid. Neurosurgery. 2007;61(5):E1101E1104. doi: 10.1227/01.neu.0000303209.38360.e6.
    1. Valdés PA, Jacobs V, Harris BT, et al. Quantitative fluorescence using 5-aminolevulinic acid-induced protoporphyrin IX biomarker as a surgical adjunct in low-grade glioma surgery. J Neurosurg. 2015;123(3):771–780. doi: 10.3171/2014.12.JNS14391.

Source: PubMed

3
Suscribir