Skip to main content
Top
Published in: Annals of Surgical Oncology 11/2018

Open Access 01-10-2018 | Pancreatic Tumors

Image-Guided Surgery in Patients with Pancreatic Cancer: First Results of a Clinical Trial Using SGM-101, a Novel Carcinoembryonic Antigen-Targeting, Near-Infrared Fluorescent Agent

Authors: Charlotte E. S. Hoogstins, MD, Leonora S. F. Boogerd, MD, Babs G. Sibinga Mulder, BSc, J. Sven D. Mieog, MD, PhD, Rutger Jan Swijnenburg, MD, PhD, Cornelis J. H. van de Velde, MD, PhD, Arantza Farina Sarasqueta, MD, PhD, Bert A. Bonsing, MD, PhD, Berenice Framery, MSc, André Pèlegrin, PhD, Marian Gutowski, MD, Françoise Cailler, PhD, Jacobus Burggraaf, MD, PhD, Alexander L. Vahrmeijer, MD, PhD

Published in: Annals of Surgical Oncology | Issue 11/2018

Login to get access

Abstract

Background

Near-infrared (NIR) fluorescence is a promising novel imaging technique that can aid in intraoperative demarcation of pancreatic cancer (PDAC) and thus increase radical resection rates. This study investigated SGM-101, a novel, fluorescent-labeled anti-carcinoembryonic antigen (CEA) antibody. The phase 1 study aimed to assess the tolerability and feasibility of intraoperative fluorescence tumor imaging using SGM-101 in patients undergoing a surgical exploration for PDAC.

Methods

At least 48 h before undergoing surgery for PDAC, 12 patients were injected intravenously with 5, 7.5, or 10 mg of SGM-101. Tolerability assessments were performed at regular intervals after dosing. The surgical field was imaged using the Quest NIR imaging system. Concordance between fluorescence and tumor presence on histopathology was studied.

Results

In this study, SGM-101 specifically accumulated in CEA-expressing primary tumors and peritoneal and liver metastases, allowing real-time intraoperative fluorescence imaging. The mean tumor-to-background ratio (TBR) was 1.6 for primary tumors and 1.7 for metastatic lesions. One false-positive lesion was detected (CEA-expressing intraductal papillary mucinous neoplasm). False-negativity was seen twice as a consequence of overlying blood or tissue that blocked the fluorescent signal.

Conclusion

The use of a fluorescent-labeled anti-CEA antibody was safe and feasible for the intraoperative detection of both primary PDAC and metastases. These results warrant further research to determine the impact of this technique on clinical decision making and overall survival.
Literature
1.
go back to reference Merkow RP, Bilimoria KY, Bentrem DJ, et al. National assessment of margin status as a quality indicator after pancreatic cancer surgery. Ann Surg Oncol. 2014;21:1067–74.CrossRef Merkow RP, Bilimoria KY, Bentrem DJ, et al. National assessment of margin status as a quality indicator after pancreatic cancer surgery. Ann Surg Oncol. 2014;21:1067–74.CrossRef
2.
go back to reference Chang DK, Johns AL, Merrett ND, et al. Margin clearance and outcome in resected pancreatic cancer. J Clin Oncol. 2009;27:2855–62.CrossRef Chang DK, Johns AL, Merrett ND, et al. Margin clearance and outcome in resected pancreatic cancer. J Clin Oncol. 2009;27:2855–62.CrossRef
3.
go back to reference Esposito I, Kleeff J, Bergmann F, et al. Most pancreatic cancer resections are R1 resections. Ann Surg Oncol. 2008;15:1651–60.CrossRef Esposito I, Kleeff J, Bergmann F, et al. Most pancreatic cancer resections are R1 resections. Ann Surg Oncol. 2008;15:1651–60.CrossRef
4.
go back to reference Garcea G, Dennison AR, Pattenden CJ, Neal CP, Sutton CD, Berry DP. Survival following curative resection for pancreatic ductal adenocarcinoma: a systematic review of the literature. JOP. 2008;9:99–132.PubMed Garcea G, Dennison AR, Pattenden CJ, Neal CP, Sutton CD, Berry DP. Survival following curative resection for pancreatic ductal adenocarcinoma: a systematic review of the literature. JOP. 2008;9:99–132.PubMed
5.
go back to reference 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;10:507–18.CrossRef 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;10:507–18.CrossRef
6.
go back to reference Hutteman M, van der Vorst JR, Mieog JS, et al. Near-infrared fluorescence imaging in patients undergoing pancreaticoduodenectomy. Eur Surg Res. 2011;47:90–7.CrossRef Hutteman M, van der Vorst JR, Mieog JS, et al. Near-infrared fluorescence imaging in patients undergoing pancreaticoduodenectomy. Eur Surg Res. 2011;47:90–7.CrossRef
7.
go back to reference 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;21:3658–66.CrossRef 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;21:3658–66.CrossRef
8.
go back to reference 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;7:392–401.CrossRef 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;7:392–401.CrossRef
9.
go back to reference Hoogstins CE, Tummers QR, Gaarenstroom KN, et 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;22:2929–38.CrossRef Hoogstins CE, Tummers QR, Gaarenstroom KN, et 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;22:2929–38.CrossRef
10.
go back to reference de Geus SW, Boogerd LS, Swijnenburg RJ, et al. Selecting tumor-specific molecular targets in pancreatic adenocarcinoma: paving the way for image-guided pancreatic surgery. Mol Imaging Biol. 2016;18:807–19.CrossRef de Geus SW, Boogerd LS, Swijnenburg RJ, et al. Selecting tumor-specific molecular targets in pancreatic adenocarcinoma: paving the way for image-guided pancreatic surgery. Mol Imaging Biol. 2016;18:807–19.CrossRef
11.
go back to reference Nap M, Mollgard K, Burtin P, Fleuren GJ. Immunohistochemistry of carcino-embryonic antigen in the embryo, fetus and adult. Tumour Biol. 1988;9:145–53.CrossRef Nap M, Mollgard K, Burtin P, Fleuren GJ. Immunohistochemistry of carcino-embryonic antigen in the embryo, fetus and adult. Tumour Biol. 1988;9:145–53.CrossRef
12.
go back to reference Hammarstrom S. The carcinoembryonic antigen (CEA) family: structures, suggested functions, and expression in normal and malignant tissues. Semin Cancer Biol. 1999;9:67–81.CrossRef Hammarstrom S. The carcinoembryonic antigen (CEA) family: structures, suggested functions, and expression in normal and malignant tissues. Semin Cancer Biol. 1999;9:67–81.CrossRef
13.
go back to reference Bacac M, Fauti T, Sam J, et al. A novel carcinoembryonic antigen T-cell bispecific antibody (CEA TCB) for the treatment of solid tumors. Clin Cancer Res. 2016;22:3286–97.CrossRef Bacac M, Fauti T, Sam J, et al. A novel carcinoembryonic antigen T-cell bispecific antibody (CEA TCB) for the treatment of solid tumors. Clin Cancer Res. 2016;22:3286–97.CrossRef
14.
go back to reference Sahlmann CO, Homayounfar K, Niessner M, et al. Repeated adjuvant anti-CEA radioimmunotherapy after resection of colorectal liver metastases: safety, feasibility, and long-term efficacy results of a prospective phase 2 study. Cancer. 2017;123:638–49.CrossRef Sahlmann CO, Homayounfar K, Niessner M, et al. Repeated adjuvant anti-CEA radioimmunotherapy after resection of colorectal liver metastases: safety, feasibility, and long-term efficacy results of a prospective phase 2 study. Cancer. 2017;123:638–49.CrossRef
15.
go back to reference Duggan MC, Jochems C, Donahue RN, et al. A phase I study of recombinant (r) vaccinia-CEA(6D)-TRICOM and rFowlpox-CEA(6D)-TRICOM vaccines with GM-CSF and IFN-alpha-2b in patients with CEA-expressing carcinomas. Cancer Immunol Immunother. 2016;65:1353–64.CrossRef Duggan MC, Jochems C, Donahue RN, et al. A phase I study of recombinant (r) vaccinia-CEA(6D)-TRICOM and rFowlpox-CEA(6D)-TRICOM vaccines with GM-CSF and IFN-alpha-2b in patients with CEA-expressing carcinomas. Cancer Immunol Immunother. 2016;65:1353–64.CrossRef
16.
go back to reference Bodet-Milin C, Ferrer L, Rauscher A, et al. Pharmacokinetics and dosimetry studies for optimization of pretargeted radioimmunotherapy in CEA-expressing advanced lung cancer patients. Front Med Lausanne. 2015;2:84.PubMedPubMedCentral Bodet-Milin C, Ferrer L, Rauscher A, et al. Pharmacokinetics and dosimetry studies for optimization of pretargeted radioimmunotherapy in CEA-expressing advanced lung cancer patients. Front Med Lausanne. 2015;2:84.PubMedPubMedCentral
17.
go back to reference Schoffelen R, Boerman OC, Goldenberg DM, et al. Development of an imaging-guided CEA-pretargeted radionuclide treatment of advanced colorectal cancer: first clinical results. Br J Cancer. 2013;109:934–42.CrossRef Schoffelen R, Boerman OC, Goldenberg DM, et al. Development of an imaging-guided CEA-pretargeted radionuclide treatment of advanced colorectal cancer: first clinical results. Br J Cancer. 2013;109:934–42.CrossRef
18.
go back to reference Gutowski M, Framery B, Boonstra MC, et al. SGM-101: an innovative near-infrared dye-antibody conjugate that targets CEA for fluorescence-guided surgery. Surg Oncol. 2017;26:153–62.CrossRef Gutowski M, Framery B, Boonstra MC, et al. SGM-101: an innovative near-infrared dye-antibody conjugate that targets CEA for fluorescence-guided surgery. Surg Oncol. 2017;26:153–62.CrossRef
19.
go back to reference Metildi CA, Kaushal S, Pu M, et al. Fluorescence-guided surgery with a fluorophore-conjugated antibody to carcinoembryonic antigen (CEA) that highlights the tumor, improves surgical resection, and increases survival in orthotopic mouse models of human pancreatic cancer. Ann Surg Oncol. 2014;21:1405–11.CrossRef Metildi CA, Kaushal S, Pu M, et al. Fluorescence-guided surgery with a fluorophore-conjugated antibody to carcinoembryonic antigen (CEA) that highlights the tumor, improves surgical resection, and increases survival in orthotopic mouse models of human pancreatic cancer. Ann Surg Oncol. 2014;21:1405–11.CrossRef
20.
go back to reference Kaushal S, McElroy M, Luiken GA, et al. Fluorophore-conjugated anti-CEA antibody for the intraoperative imaging of pancreatic and colorectal cancer. J Gastrointest Surg. 2008;12:1938–50.CrossRef Kaushal S, McElroy M, Luiken GA, et al. Fluorophore-conjugated anti-CEA antibody for the intraoperative imaging of pancreatic and colorectal cancer. J Gastrointest Surg. 2008;12:1938–50.CrossRef
21.
go back to reference van Driel PB, van de Giessen M, Boonstra MC, et al. Characterization and evaluation of the artemis camera for fluorescence-guided cancer surgery. Mol Imaging Biol. 2015;17:413–23.CrossRef van Driel PB, van de Giessen M, Boonstra MC, et al. Characterization and evaluation of the artemis camera for fluorescence-guided cancer surgery. Mol Imaging Biol. 2015;17:413–23.CrossRef
22.
go back to reference Frangioni JV. New technologies for human cancer imaging. J Clin Oncol. 2008;26:4012–21.CrossRef Frangioni JV. New technologies for human cancer imaging. J Clin Oncol. 2008;26:4012–21.CrossRef
23.
go back to reference Chance B. Near-infrared images using continuous, phase-modulated, and pulsed light with quantitation of blood and blood oxygenation. Ann N Y Acad Sci. 1998;838:29–45.CrossRef Chance B. Near-infrared images using continuous, phase-modulated, and pulsed light with quantitation of blood and blood oxygenation. Ann N Y Acad Sci. 1998;838:29–45.CrossRef
24.
go back to reference Hekman MC, Boerman OC, de Weijert M, et al. Targeted dual-modality imaging in renal cell carcinoma: an ex vivo kidney perfusion study. Clin Cancer Res. 2016;22:4634–42.CrossRef Hekman MC, Boerman OC, de Weijert M, et al. Targeted dual-modality imaging in renal cell carcinoma: an ex vivo kidney perfusion study. Clin Cancer Res. 2016;22:4634–42.CrossRef
25.
go back to reference Chiorean EG, Coveler AL. Pancreatic cancer: optimizing treatment options, new, and emerging targeted therapies. Drug Des Devel Ther. 2015;9:29–3545. Chiorean EG, Coveler AL. Pancreatic cancer: optimizing treatment options, new, and emerging targeted therapies. Drug Des Devel Ther. 2015;9:29–3545.
26.
go back to reference Boonstra MC, Prakash J, Van De Velde CJ, et al. Stromal targets for fluorescent-guided oncologic surgery. Front Oncol. 2015;5:254.CrossRef Boonstra MC, Prakash J, Van De Velde CJ, et al. Stromal targets for fluorescent-guided oncologic surgery. Front Oncol. 2015;5:254.CrossRef
27.
go back to reference Tipirneni KE, Warram JM, Moore LS, et al. Oncologic procedures amenable to fluorescence-guided surgery. Ann Surg. 2017;266:36–47.CrossRef Tipirneni KE, Warram JM, Moore LS, et al. Oncologic procedures amenable to fluorescence-guided surgery. Ann Surg. 2017;266:36–47.CrossRef
Metadata
Title
Image-Guided Surgery in Patients with Pancreatic Cancer: First Results of a Clinical Trial Using SGM-101, a Novel Carcinoembryonic Antigen-Targeting, Near-Infrared Fluorescent Agent
Authors
Charlotte E. S. Hoogstins, MD
Leonora S. F. Boogerd, MD
Babs G. Sibinga Mulder, BSc
J. Sven D. Mieog, MD, PhD
Rutger Jan Swijnenburg, MD, PhD
Cornelis J. H. van de Velde, MD, PhD
Arantza Farina Sarasqueta, MD, PhD
Bert A. Bonsing, MD, PhD
Berenice Framery, MSc
André Pèlegrin, PhD
Marian Gutowski, MD
Françoise Cailler, PhD
Jacobus Burggraaf, MD, PhD
Alexander L. Vahrmeijer, MD, PhD
Publication date
01-10-2018
Publisher
Springer International Publishing
Published in
Annals of Surgical Oncology / Issue 11/2018
Print ISSN: 1068-9265
Electronic ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-018-6655-7

Other articles of this Issue 11/2018

Annals of Surgical Oncology 11/2018 Go to the issue