1
|
Qiu SQ, He XF, Liang XL, Shi GY, Zhao ML, Li F, Wu ZY, Tian J, Zhai TT, Du Y. GLUT1 as a generic biomarker enables near-infrared fluorescence molecular imaging guided precise intraoperative tumor detection in breast cancer. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07095-4. [PMID: 39833507 DOI: 10.1007/s00259-025-07095-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE Precise tumor excision is important but challenging in breast-conserving surgery (BCS). Tumor-specific fluorescence imaging may be used for intraoperative tumor detection and, therefore, to guide precise tumor excision. The aims of this study are to develop a glucose transporter 1 (GLUT1)-targeted near-infrared fluorescence tracer and evaluate its accuracy for breast cancer detection using fresh surgical breast specimens. METHODS Bioinformatic analysis was performed to compare GLUT1 expression between breast cancer and normal breast tissues. A GLUT1-targeted fluorescence imaging tracer WZB117-CY7.5 was developed. In combination with fluorescence imaging (FMI), its binding specificity to GLUT1 was examined in in vitro breast cancer cell experiments, in vivo 4T1 breast tumor-bearing mouse models, and 60 freshly resected human breast tumor tissues. The diagnostic accuracy of WZB117-CY7.5, was evaluated in fresh specimens derived from 60 patients diagnosed with breast cancer. RESULTS GLUT1 expression is higher in breast cancer tissues compared with normal tissues. WZB117-CY7.5 specifically bound to breast cancer cells in in vitro cell experiments and accumulated in tumor areas in a 4T1 tumor-bearing mice after intravenous injection by FMI. Moreover, WZB117-CY7.5 specifically bound to freshly resected human breast cancer and demonstrated excellent diagnostic performance in discriminating breast cancer, irrespective of cancer subtype, from normal breast tissue on fresh surgically resected breast tissues. CONCLUSIONS WZB117-CY7.5 showed high accuracy in intraoperative breast cancer detection, irrespective of the cancer subtype. This highlights its potential for clinical applications as a generic tracer for fluorescence image-guided surgery (FIGS) in BCS and fluorescence image-guided pathology for tissue sampling.
Collapse
Affiliation(s)
- Si-Qi Qiu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, No.114 Waima Road, Shantou, 515041, Shantou, China
- Clinical Research Center, Shantou Central Hospital, Shantou, 515041, China
| | - Xiao-Feng He
- Shantou University Medical College, Shantou, 515041, China
| | - Xiao-Long Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Guang-Yuan Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China
| | - Meng-Long Zhao
- Clinical Research Center, Shantou Central Hospital, Shantou, 515041, China
| | - Fan Li
- Biobank, Shantou Central Hospital, Shantou, 515041, China
| | - Zhi-Yong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, No.114 Waima Road, Shantou, 515041, Shantou, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China.
- Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100191, China.
| | - Tian-Tian Zhai
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, No.7 Raoping Road, Jinping District, Shantou, 515041, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, No.95 Zhongguancun East Road, Hai Dian District, Beijing, 100190, China.
- The University of Chinese Academy of Sciences, Beijing, 100080, China.
| |
Collapse
|
2
|
Keizers B, Nijboer TS, van der Fels CAM, van den Heuvel MC, van Dam GM, Kruijff S, Jan de Jong I, Witjes MJH, Voskuil FJ, Gorpas D, Browne WR, van der Zaag PJ. Systematic comparison of fluorescence imaging in the near-infrared and shortwave-infrared spectral range using clinical tumor samples containing cetuximab-IRDye800CW. JOURNAL OF BIOMEDICAL OPTICS 2025; 30:S13708. [PMID: 39553388 PMCID: PMC11566260 DOI: 10.1117/1.jbo.30.s1.s13708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/25/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Significance Shortwave-infrared (SWIR) imaging is reported to yield better contrast in fluorescence-guided surgery than near-infrared (NIR) imaging, due to a reduction in scattering. This benefit of SWIR was shown in animal studies, however not yet in clinical studies with patient samples. Aim We investigate the potential benefit of SWIR to NIR imaging in clinical samples containing cetuximab-IRDye800CW in fluorescence-guided surgery. Approach The potential of the epidermal growth factor-targeted NIR dye cetuximab-IRDye800CW in the shortwave range was examined by recording the absorption and emission spectrum. An ex vivo comparison of NIR and SWIR images using clinical tumor samples of patients with penile squamous cell carcinoma (PSCC) and head and neck squamous cell carcinoma (HNSCC) containing cetuximab-IRDye800CW was performed. The comparison was based on the tumor-to-background ratio and an adapted contrast-to-noise ratio (aCNR) using the standard of care pathology tissue assessment as the golden standard. Results Based on the emission spectrum, cetuximab-IRDye800CW can be detected in the SWIR range. In clinical PSCC samples, overall SWIR imaging was found to perform similarly to NIR imaging (NIR imaging is better than SWIR in the 2/7 criteria examined, and SWIR is better than NIR in the 3/7 criteria). However, when inspecting HNSCC data, NIR is better than SWIR in nearly all (5/7) examined criteria. This difference seems to originate from background autofluorescence overwhelming the off-peak SWIR fluorescence signal in HNSCC tissue. Conclusion SWIR imaging using the targeted tracer cetuximab-IRDye800CW currently does not provide additional benefit over NIR imaging in ex vivo clinical samples. Background fluorescence in the SWIR region, resulting in a higher background signal, limits SWIR imaging in HNSCC samples. However, SWIR shows potential in increasing the contrast of tumor borders in PSCC samples, as shown by a higher aCNR over a line.
Collapse
Affiliation(s)
- Bas Keizers
- University Medical Centre Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Surgery, Groningen, The Netherlands
| | - Thomas S. Nijboer
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | | | - Marius C. van den Heuvel
- University Medical Centre Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | | | - Schelto Kruijff
- University Medical Centre Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Surgery, Groningen, The Netherlands
| | - Igle Jan de Jong
- University Medical Centre Groningen, Department of Urology, Groningen, The Netherlands
| | - Max J. H. Witjes
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Floris J. Voskuil
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University Medical Centre Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Dimitris Gorpas
- Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany
- Technische Universität München, School of Medicine and Health, Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), München, Germany
| | - Wesley R. Browne
- University of Groningen, Stratingh Institute for Chemistry, Faculty of Science and Engineering, Groningen, The Netherlands
| | - Pieter J. van der Zaag
- University Medical Centre Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
- University Medical Centre Groningen, Optical Molecular Imaging Groningen, Groningen, The Netherlands
- University of Groningen, Zernike Institute, Molecular Biophysics, Groningen, The Netherlands
| |
Collapse
|
3
|
Gupta S, Pal R, Schmidt EJ, Krishnamoorthy M, Leporati A, Kumar AT, Bogdanov A. Miniaturized Fab' imaging probe derived from a clinical antibody: Characterization and imaging in CRISPRi-attenuated mammary tumor models. iScience 2024; 27:110102. [PMID: 39184438 PMCID: PMC11342199 DOI: 10.1016/j.isci.2024.110102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/28/2024] [Accepted: 05/22/2024] [Indexed: 08/27/2024] Open
Abstract
Clinical imaging-assisted oncosurgical navigation requires cancer-specific miniaturized optical imaging probes. We report a near-infrared (NIR) Fab'-based epidermal growth factor receptor (EGFR)-specific probe carrying 3 NIR fluorophores (Fab'-800CW), which retained high-affinity binding to EGFR ectodomain (equilibrium KD E = 1 nM). Fab'-800CW showed a robust 4-times gain of fluorescence intensity (FI) and a 20% lifetime (FLT) increase under the conditions mimicking intracellular degradation. The probe was tested by using triple-negative breast cancer (TNBC) cell lines obtained by applying CRISPR interference (CRISPRi) effect of EGFR-targeting sgRNA and dCas9-KRAB chimera coexpression in MDA-MB-231 cells (WT cells). FI imaging in cell culture proved a 50% EGFR expression attenuation by CRISPRi. FI imaging in animals harboring attenuated or WT TNBC tumors with ex vivo corroboration identified differences between WT and CRISPRi tumors FI at 30 min post injection. Our results suggest the feasibility of EGFR expression imaging using a Fab'-based probe relevant for imaging-guided cancer surgery.
Collapse
Affiliation(s)
- Suresh Gupta
- Department of Radiology, UMASS Chan Medical School, Worcester, MA, USA
| | - Rahul Pal
- Mike Toth Head and Neck Cancer Research Center, Massachusetts Eye and Ear, Boston, MA, USA
- A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Eric J. Schmidt
- Department of Radiology, UMASS Chan Medical School, Worcester, MA, USA
| | - Murali Krishnamoorthy
- Mike Toth Head and Neck Cancer Research Center, Massachusetts Eye and Ear, Boston, MA, USA
- A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Anita Leporati
- Department of Radiology, UMASS Chan Medical School, Worcester, MA, USA
| | - Anand T.N. Kumar
- Mike Toth Head and Neck Cancer Research Center, Massachusetts Eye and Ear, Boston, MA, USA
- A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Alexei Bogdanov
- Department of Radiology, UMASS Chan Medical School, Worcester, MA, USA
- Cancer Center and Chemical Biology Interface Program, UMASS Chan Medical School, Worcester, MA, USA
| |
Collapse
|
4
|
Nijboer TS, van der Fels CAM, de Wit JG, Keizers B, Huizinga HK, Voskuil FJ, Voskamp MJH, van den Heuvel MC, Witjes MJH, de Jong IJ. Fluorescence-guided surgery using cetuximab-800CW in patients with penile carcinoma. BJU Int 2024; 134:268-275. [PMID: 38659306 DOI: 10.1111/bju.16384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
OBJECTIVE To investigate the feasibility of fluorescence molecular imaging (FMI), using cetuximab-800CW, as an intraoperative tool to determine surgical margins in penile squamous cell carcinoma (PSCC). PATIENTS AND METHODS A total of 11 patients with PSCC received 75 mg cetuximab followed by 15 mg cetuximab-800CW 2 days before surgery. FMI of the whole excision specimen and tissue slices was performed. Fluorescence visualisation was correlated to histopathology. Based on tumour and healthy tissue regions of interest, mean fluorescence intensity was calculated for each individual patient. RESULTS Significant differences between tumour and healthy mean fluorescence intensity were found with tumour-to-background ratios of a median (IQR) of 1.51 (0.99) and a mean (SD) of 1.51 (0.32) in the excision specimen and tissue slices, respectively. One patient showed a high relative fluorescence intensity with a signal-to-background ratio of 1.79, corresponding to a tumour-positive margin on fresh frozen sectioning. CONCLUSION In this Phase I study we showed that cetuximab-800CW seems suitable to discriminate PSCC from background tissue. The tracer was well tolerated, and no false positive spots were seen.
Collapse
Affiliation(s)
- Thomas S Nijboer
- Department of Oral and Maxillofacial Surgery, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Jaron G de Wit
- Department of Oral and Maxillofacial Surgery, University Medical Centre Groningen, Groningen, The Netherlands
| | - Bas Keizers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, Groningen, The Netherlands
| | - Henrik K Huizinga
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Floris J Voskuil
- Department of Oral and Maxillofacial Surgery, University Medical Centre Groningen, Groningen, The Netherlands
| | - Maarten J H Voskamp
- Department of Urology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Marius C van den Heuvel
- Department of Pathology and Medical Biology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Max J H Witjes
- Department of Oral and Maxillofacial Surgery, University Medical Centre Groningen, Groningen, The Netherlands
| | - Igle Jan de Jong
- Department of Urology, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
5
|
Rounds CC, de Wit JG, Vonk J, Vorjohan J, Nelson S, Trang A, Villinski B, Samkoe KS, Brankov JG, Voskuil FJ, Witjes MJH, Tichauer KM. Improved intraoperative identification of close margins in oral squamous cell carcinoma resections using a dual aperture fluorescence ratio approach: first in-human results. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:016003. [PMID: 38235321 PMCID: PMC10793906 DOI: 10.1117/1.jbo.29.1.016003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024]
Abstract
Significance Surgical excision is the main treatment for solid tumors in oral squamous cell carcinomas, where wide local excision (achieving a healthy tissue margin of > 5 mm around the excised tumor) is the goal as it results in reduced local recurrence rates and improved overall survival. Aim No clinical methods are available to assess the complete surgical margin intraoperatively while the patient is still on the operating table; and while recent intraoperative back-bench fluorescence-guided surgery approaches have shown promise for detecting "positive" inadequate margins (< 1 mm ), they have had limited success in the detection of "close" inadequate margins (1 to 5 mm). Here, a dual aperture fluorescence ratio (dAFR) approach was evaluated as a means of improving detection of close margins. Approach The approach was evaluated on surgical specimens from patients who were administered a tumor-specific fluorescent imaging agent (cetuximab-800CW) prior to surgery. The dAFR approach was compared directly against standard wide-field fluorescence imaging and pathology measurements of margin thickness in specimens from three patients and a total of 12 margin locations (1 positive, 5 close, and 6 clear margins). Results The area under the receiver operating characteristic curve, representing the ability to detect close compared to clear margins (> 5 mm ) was found to be 1.0 and 0.57 for dAFR and sAF, respectively. Improvements in dAFR were found to be statistically significant (p < 0.02 ). Conclusions These results provide evidence that the dAFR approach potentially improves detection of close surgical margins.
Collapse
Affiliation(s)
- Cody C. Rounds
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Jaron G. de Wit
- University Medical Center Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Jasper Vonk
- University Medical Center Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
- University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
| | - Jennifer Vorjohan
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Sophia Nelson
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Allyson Trang
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Brooke Villinski
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Kimberley S. Samkoe
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Jovan G. Brankov
- University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
- Illinois Institute of Technology, Department of Electrical and Computer Engineering, Chicago Illinois, United States
| | - Floris J. Voskuil
- University Medical Center Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Max J. H. Witjes
- University Medical Center Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Kenneth M. Tichauer
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| |
Collapse
|
6
|
de Wit JG, Vonk J, Voskuil FJ, de Visscher SAHJ, Schepman KP, Hooghiemstra WTR, Linssen MD, Elias SG, Halmos GB, Plaat BEC, Doff JJ, Rosenthal EL, Robinson D, van der Vegt B, Nagengast WB, van Dam GM, Witjes MJH. EGFR-targeted fluorescence molecular imaging for intraoperative margin assessment in oral cancer patients: a phase II trial. Nat Commun 2023; 14:4952. [PMID: 37587149 PMCID: PMC10432510 DOI: 10.1038/s41467-023-40324-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 07/21/2023] [Indexed: 08/18/2023] Open
Abstract
Inadequate surgical margins occur frequently in oral squamous cell carcinoma surgery. Fluorescence molecular imaging (FMI) has been explored for intraoperative margin assessment, but data are limited to phase-I studies. In this single-arm phase-II study (NCT03134846), our primary endpoints were to determine the sensitivity, specificity and positive predictive value of cetuximab-800CW for tumor-positive margins detection. Secondary endpoints were safety, close margin detection rate and intrinsic cetuximab-800CW fluorescence. In 65 patients with 66 tumors, cetuximab-800CW was well-tolerated. Fluorescent spots identified in the surgical margin with signal-to-background ratios (SBR) of ≥2 identify tumor-positive margins with 100% sensitivity, 85.9% specificity, 58.3% positive predictive value, and 100% negative predictive value. An SBR of ≥1.5 identifies close margins with 70.3% sensitivity, 76.1% specificity, 60.5% positive predictive value, and 83.1% negative predictive value. Performing frozen section analysis aimed at the fluorescent spots with an SBR of ≥1.5 enables safe, intraoperative adjustment of surgical margins.
Collapse
Affiliation(s)
- Jaron G de Wit
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Jasper Vonk
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Floris J Voskuil
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Pathology & Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Sebastiaan A H J de Visscher
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Kees-Pieter Schepman
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Wouter T R Hooghiemstra
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Matthijs D Linssen
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gyorgy B Halmos
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Boudewijn E C Plaat
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Jan J Doff
- Department of Pathology & Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Eben L Rosenthal
- Department of Otolaryngology, Vanderbilt University Medical Centre, Nashville, Tennessee, United States of America
| | - Dominic Robinson
- Center for Optical Diagnostics and Therapy, Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Bert van der Vegt
- Department of Pathology & Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Wouter B Nagengast
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Gooitzen M van Dam
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- TRACER Europe B.V. / AxelaRx, Groningen, the Netherlands
| | - Max J H Witjes
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands.
| |
Collapse
|
7
|
Sikkenk DJ, Sterkenburg AJ, Schmidt I, Gorpas D, Nagengast WB, Consten ECJ. Detection of Tumour-Targeted IRDye800CW Tracer with Commercially Available Laparoscopic Surgical Systems. Diagnostics (Basel) 2023; 13:diagnostics13091591. [PMID: 37174982 PMCID: PMC10178288 DOI: 10.3390/diagnostics13091591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
(1) Introduction: Near-infrared fluorescence (NIRF) combined with tumour-targeted tracers, such as bevacizumab-800CW, could aid surgical decision-making. This study explored the use of IRDye800CW, conjugated to bevacizumab, with four commercially available NIRF laparoscopes optimised for indocyanine green (ICG). (2) Methods: A (lymph node) phantom was made from a calibration device for NIRF and tissue-mimicking material. Serial dilutions of bevacizumab-800CW were made and ICG functioned as a reference. System settings, working distance, and thickness of tissue-mimicking material were varied to assess visibility of the fluorescence signal and tissue penetration. Tests were performed with four laparoscopes: VISERA ELITE II, Olympus; IMAGE1 S™ 4U Rubina, KARL STORZ; ENDOCAM Logic 4K platform, Richard Wolf; da Vinci Xi, Intuitive Surgical. (3) Results: The lowest visible bevacizumab-800CW concentration ranged between 13-850 nM (8-512 times diluted stock solution) for all laparoscopes, but the tracer was not visible through 0.8 cm of tissue in all systems. In contrast, ICG was still visible at a concentration of 0.4 nM (16,384 times diluted) and through 1.6-2.4 cm of tissue. Visibility and tissue penetration generally improved with a reduced working distance and manually adjusted system settings. (4) Conclusion: Depending on the application, bevacizumab-800CW might be sufficiently visible with current laparoscopes, but optimisation would widen applicability of tumour-targeted IRDye800CW tracers.
Collapse
Affiliation(s)
- Daan J Sikkenk
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Surgery, Meander Medical Centre, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| | - Andrea J Sterkenburg
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Dimitris Gorpas
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, Ismaninger Straße 22, D-81675 Munich, Germany
| | - Wouter B Nagengast
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Esther C J Consten
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Surgery, Meander Medical Centre, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| |
Collapse
|
8
|
Wang C, Xu X, Folaron M, Gunn JR, Hodge S, Chen EY, Hoopes PJ, Tichauer KM, Samkoe KS. Improved Discrimination of Tumors with Low and Heterogeneous EGFR Expression in Fluorescence-Guided Surgery Through Paired-Agent Protocols. Mol Imaging Biol 2023; 25:110-121. [PMID: 34651290 PMCID: PMC9527767 DOI: 10.1007/s11307-021-01656-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/16/2021] [Accepted: 07/18/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE The goal of fluorescence-guided surgery (FGS) in oncology is to improve the surgical therapeutic index by enhancing contrast between cancerous and healthy tissues. However, optimal discrimination between these tissues is complicated by the nonspecific uptake and retention of molecular targeted agents and the variance of fluorescence signal. Paired-agent imaging (PAI) employs co-administration of an untargeted imaging agent with a molecular targeted agent, providing a normalization factor to minimize nonspecific and varied signals. The resulting measured binding potential is quantitative and equivalent to in vivo immunohistochemistry of the target protein. This study demonstrates that PAI improves the accuracy of tumor-to-healthy tissue discrimination compared to single-agent imaging for in vivo FGS. PROCEDURES PAI using a fluorescent anti-epidermal growth factor receptor (EGFR) affibody molecule (ABY-029, eIND 122,681) with untargeted IRDye 700DX carboxylate was compared to ABY-029 alone in an oral squamous cell carcinoma xenograft mouse model at 3 h after dye administration (n = 30). RESULTS PAI significantly enhanced tumor discrimination, as compared to ABY-029 alone in low EGFR-expressing tumors and highly heterogeneous populations including multiple cell lines with varying expression (diagnostic accuracy: 0.908 vs. 0.854 and 0.908 vs. 0.822; and ROC curve AUC: 0.963 vs. 0.909 and 0.957 vs. 0.909, respectively) indicating a potential for universal FGS image thresholds to determine surgical margins. In addition, PAI achieved significantly higher diagnostic ability than ABY-029 alone 0.25-5-h post injection and exhibited a stronger correlation to EGFR expression heterogeneity. CONCLUSION The quantitative receptor delineation of PAI promises to improve the surgical therapeutic index of cancer resection in a clinically relevant timeline.
Collapse
Affiliation(s)
- Cheng Wang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Xiaochun Xu
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Margaret Folaron
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Jason R Gunn
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Sassan Hodge
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Eunice Y Chen
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - P Jack Hoopes
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Kenneth M Tichauer
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Kimberley S Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
9
|
Li C, Mi J, Wang Y, Zhang Z, Guo X, Zhou J, Hu Z, Tian J. New and effective EGFR-targeted fluorescence imaging technology for intraoperative rapid determination of lung cancer in freshly isolated tissue. Eur J Nucl Med Mol Imaging 2023; 50:494-507. [PMID: 36207638 DOI: 10.1007/s00259-022-05975-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/15/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE During lung cancer surgery, it is very important to define tumor boundaries and determine the surgical margin distance. In previous research, systemically application of fluorescent probes can help medical professionals determine the boundaries of tumors and find small tumors and metastases, thereby improving the accuracy of surgical resection. However, there are very few safe and effective probes that can be applied to clinical trials up to now, which limits the clinical application of fluorescence imaging. Here we developed a new technology that can quickly identify the tumor area in the resected lung tissue during the operation and distinguish the tumor boundary and metastatic lymph nodes. EXPERIMENTAL DESIGN For animal studies, a PDX model of lung cancer was established. The tumors, lungs, and peritumoral muscle tissues of tumor-bearing mice were surgically removed and incubated with a probe targeting epidermal growth factor receptor (EGFR) for 20 min, and then imaged by a closed-field near-infrared two-zone (NIR-II) fluorescence imaging system. For clinical samples, ten surgically removed lung tissues and 60 lymph nodes from 10 lung cancer patients undergoing radical resection were incubated with the targeting probe immediately after intraoperative resection and imaged to identify the tumor area and distinguish the tumor boundary and metastatic lymph nodes. The accuracy of fluorescence imaging was confirmed by HE staining and immunohistochemistry. RESULTS The ex vivo animal imaging experiments showed a fluorescence enhancement of tumor tissue. For clinical samples, our results showed that this new technology yielded more than 85.7% sensitivity and 100% specificity in identifying the tumor area in the resected lung tissue. The average fluorescence tumor-to-background ratio was 2.5 ± 1.3. Furthermore, we also used this technique to image metastatic lymph nodes intraoperatively and showed that metastatic lymph nodes have brighter fluorescence than normal lymph nodes, as the average fluorescence tumor-to-background signal ratio was 2.7 ± 1.1. Calculations on the results of the fluorescence signal in relation to the number of metastatic lymph nodes yielded values of 77.8% for sensitivity and 92.1% for specificity. We expect this new technology to be a useful diagnostic tool for rapid intraoperative pathological detection and margin determination. CONCLUSIONS By using fluorescently labeled anti-human EGFR recombinant antibody scFv fragment to incubate freshly isolated tissues during surgery, the probes can quickly accumulate in lung cancer tissues, which can be used to quickly identify tumor areas in the resected lung tissues and distinguish tumor boundaries and find metastases in lymph nodes. This technology is expected to be used for rapid intraoperative pathological detection and margin determination.
Collapse
Affiliation(s)
- Changjian Li
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, 100191, People's Republic of China
| | - Jiahui Mi
- Department of Thoracic Surgery, Peking University People's Hospital, No.11, Xi Zhi Men South Avenue, Beijing, 100190, People's Republic of China
| | - Yueqi Wang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Zeyu Zhang
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, 100191, People's Republic of China
| | - Xiaoyong Guo
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, People's Republic of China
- Department of Gastroenterology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, 100190, People's Republic of China
| | - Jian Zhou
- Department of Thoracic Surgery, Peking University People's Hospital, No.11, Xi Zhi Men South Avenue, Beijing, 100190, People's Republic of China.
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, People's Republic of China.
| | - Jie Tian
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, 100191, People's Republic of China.
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, People's Republic of China.
- Zhuhai Precision Medical Center, Zhuhai People's Hospital, Affiliated With Jinan University, Zhuhai, 519000, People's Republic of China.
| |
Collapse
|
10
|
de Wit JG, van Schaik JE, Voskuil FJ, Vonk J, de Visscher SAHJ, Schepman KP, van der Laan BFAM, Doff JJ, van der Vegt B, Plaat BEC, Witjes MJH. Comparison of narrow band and fluorescence molecular imaging to improve intraoperative tumour margin assessment in oral cancer surgery. Oral Oncol 2022; 134:106099. [PMID: 36096045 DOI: 10.1016/j.oraloncology.2022.106099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/04/2022] [Accepted: 08/23/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE New techniques have emerged to aid in preventing inadequate margins in oral squamous cell carcinoma (OSCC) surgery, but studies comparing different techniques are lacking. Here, we compared narrow band imaging (NBI) with fluorescence molecular imaging (FMI), to study which intraoperative technique best assesses the mucosal tumour margins. MATERIALS AND METHODS NBI was performed in vivo and borders were marked with three sutures. For FMI, patients received 75 mg of unlabelled cetuximab followed by 15 mg cetuximab-800CW intravenously-two days prior to surgery. The FMI borders were defined on the excised specimen. The NBI borders were correlated with the FMI outline and histopathology. RESULTS Sixteen patients were included, resulting in 31 NBI and 30 FMI measurements. The mucosal border was delineated within 1 mm of the tumour border in 4/31 (13 %) of NBI and in 16/30 (53 %) FMI cases (p = 0.0008), and within 5 mm in 23/31 (74 %) of NBI and in 29/30 (97 %) of FMI cases (p = 0.0048). The median distance between the tumour border and the imaging border was significantly greater for NBI (3.2 mm, range -6.1 to 12.8 mm) than for FMI (0.9 mm, range -3.0 to 7.4 mm; p = 0.028). Submucosal extension and previous irradiation reduced NBI accuracy. CONCLUSION Ex vivo FMI performed more accurately than in vivo NBI in mucosal margin assessment, mainly because NBI cannot detect submucosal extension. NBI adequately identified the mucosal margin especially in early-stage and not previously irradiated tumours, and may therefore be preferable in these tumours for practical and cost-related reasons.
Collapse
Affiliation(s)
- Jaron G de Wit
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Jeroen E van Schaik
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Floris J Voskuil
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands; Department of Pathology & Medical Biology, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Jasper Vonk
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Sebastiaan A H J de Visscher
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Kees-Pieter Schepman
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Bernard F A M van der Laan
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands; Department of Otorhinolaryngology, Head and Neck Surgery, Haaglanden Medical Centre, The Hague, the Netherlands
| | - Jan J Doff
- Department of Pathology & Medical Biology, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Bert van der Vegt
- Department of Pathology & Medical Biology, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Boudewijn E C Plaat
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands
| | - Max J H Witjes
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, the Netherlands.
| |
Collapse
|
11
|
Vergeer RA, Theunissen REP, van Elk T, Schmidt I, Postma MR, Tamasi K, van Dijk JMC, Kuijlen JMA. Fluorescence-guided detection of pituitary neuroendocrine tumor (PitNET) tissue during endoscopic transsphenoidal surgery available agents, their potential, and technical aspects. Rev Endocr Metab Disord 2022; 23:647-657. [PMID: 35344185 PMCID: PMC9156450 DOI: 10.1007/s11154-022-09718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 10/29/2022]
Abstract
Differentiation of pituitary neuroendocrine tumor (PitNET) tissue from surrounding normal tissue during surgery is challenging. A number of fluorescent agents is available for visualization of tissue discrepancy, with the potential of improving total tumor resection. This review evaluates the availability, clinical and technical applicability of the various fluorescent agents within the field of pituitary surgery. According to PRISMA guidelines, a systematic review was performed to identify reports describing results of in vivo application of fluorescent agents. In this review, 15 publications were included. Sodium Fluorescein (FNa) was considered in two studies. The first study reported noticeable fluorescence in adenoma tissue, the second demonstrated the strongest fluorescence in non-functioning pituitary adenomas. 5-Aminolevulinic acid (5-ALA) was investigated in three studies. One study compared laser-based optical biopsy system (OBS) with photo-diagnostic filter (PD) and found that the OBS was able to detect all microadenomas, even when MRI was negative. The second study retrospectively analyzed twelve pituitary adenomas and found only one positive for fluorescence. The third investigated fifteen pituitary adenomas of which one displayed vague fluorescence. Indocyanine green (ICG) was researched in four studies with variable results. Second-Window ICG yielded no significant difference between functioning and non-functioning adenomas in one study, while a second study displayed 4 times higher fluorescence in tumor tissue than in normal tissue. In three studies, OTL38 showed potential in non-functioning pituitary adenomas. At present, evidence for fluorescent agents to benefit total resection of PitNETs is lacking. OTL38 can potentially serve as a selective fluorescent agent in non-functioning pituitary adenomas in the near future.
Collapse
Affiliation(s)
- Rob A Vergeer
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Robin E P Theunissen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Theodora van Elk
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mark R Postma
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Katalin Tamasi
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J Marc C van Dijk
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jos M A Kuijlen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
12
|
Vonk J, Kukačka J, Steinkamp P, de Wit J, Voskuil F, Hooghiemstra W, Bader M, Jüstel D, Ntziachristos V, van Dam G, Witjes M. Multispectral optoacoustic tomography for in vivo detection of lymph node metastases in oral cancer patients using an EGFR-targeted contrast agent and intrinsic tissue contrast: A proof-of-concept study. PHOTOACOUSTICS 2022; 26:100362. [PMID: 35541024 PMCID: PMC9079001 DOI: 10.1016/j.pacs.2022.100362] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/07/2022] [Accepted: 04/27/2022] [Indexed: 05/09/2023]
Abstract
Oral cancer patients undergo diagnostic surgeries to detect occult lymph node metastases missed by preoperative structural imaging techniques. Reducing these invasive procedures that are associated with considerable morbidity, requires better preoperative detection. Multispectral optoacoustic tomography (MSOT) is a rapidly evolving imaging technique that may improve preoperative detection of (early-stage) lymph node metastases, enabling the identification of molecular changes that often precede structural changes in tumorigenesis. Here, we characterize the optoacoustic properties of cetuximab-800CW, a tumor-specific fluorescent tracer showing several photophysical properties that benefit optoacoustic signal generation. In this first clinical proof-of-concept study, we explore its use as optoacoustic to differentiate between malignant and benign lymph nodes. We characterize the appearance of malignant lymph nodes and show differences in the distribution of intrinsic chromophores compared to benign lymph nodes. In addition, we suggest several approaches to improve the efficiency of follow-up studies.
Collapse
Affiliation(s)
- J. Vonk
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, the Netherlands
| | - J. Kukačka
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - P.J. Steinkamp
- Department of Surgery, University of Groningen, University Medical Center Groningen, the Netherlands
| | - J.G. de Wit
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, the Netherlands
| | - F.J. Voskuil
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, the Netherlands
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - W.T.R. Hooghiemstra
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M. Bader
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - D. Jüstel
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - V. Ntziachristos
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - G.M. van Dam
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- AxelaRx / TRACER B.V., Groningen, the Netherlands
| | - M.J.H. Witjes
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, the Netherlands
- Correspondence to: Department of Oral & Maxillofacial Surgery, University Medical Center Groningen, the Netherlands.
| |
Collapse
|
13
|
Multimodal CEA-targeted fluorescence and radioguided cytoreductive surgery for peritoneal metastases of colorectal origin. Nat Commun 2022; 13:2621. [PMID: 35551444 PMCID: PMC9098887 DOI: 10.1038/s41467-022-29630-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 03/16/2022] [Indexed: 11/09/2022] Open
Abstract
In patients with colorectal peritoneal metastases scheduled for cytoreductive surgery, accurate preoperative estimation of tumor burden and subsequent intraoperative detection of all tumor deposits remains challenging. In this study (ClinicalTrials.gov NCT03699332) we describe the results of a phase I clinical trial evaluating [111In]In-DOTA-labetuzumab-IRDye800CW, a dual-labeled anti-carcinoembryonic antigen (anti-CEA) antibody conjugate that enables both preoperative imaging and intraoperative radioguidance and fluorescence imaging. Primary study outcomes are safety and feasibility of this multimodal imaging approach. Secondary outcomes are determination of the optimal dose, correlation between tracer uptake and histopathology and effects on clinical strategy. Administration of [111In]In-DOTA-labetuzumab-IRDye800CW is well-tolerated and enables sensitive pre- and intraoperative imaging in patients who receive 10 or 50 mg of the tracer. Preoperative imaging revealed previously undetected lymph node metastases in one patient, and intraoperative fluorescence imaging revealed four previously undetected metastases in two patients. Alteration of clinical strategy based on multimodal imaging occurred in three patients. Thus, multimodal image-guided surgery after administration of this dual-labeled tracer is a promising approach that may aid in decision making before and during cytoreductive surgical procedures.
Collapse
|
14
|
Fluorescence grid analysis for the evaluation of piecemeal surgery in sinonasal inverted papilloma: a proof-of-concept study. Eur J Nucl Med Mol Imaging 2021; 49:1640-1649. [PMID: 34738141 PMCID: PMC8940828 DOI: 10.1007/s00259-021-05567-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/17/2021] [Indexed: 12/14/2022]
Abstract
Purpose Local recurrence occurs in ~ 19% of sinonasal inverted papilloma (SNIP) surgeries and is strongly associated with incomplete resection. During surgery, it is technically challenging to visualize and resect all SNIP tissue in this anatomically complex area. Proteins that are overexpressed in SNIP, such as vascular endothelial growth factor (VEGF), may serve as a target for fluorescence molecular imaging to guide surgical removal of SNIP. A proof-of-concept study was performed to investigate if the VEGF-targeted near-infrared fluorescent tracer bevacizumab-800CW specifically localizes in SNIP and whether it could be used as a clinical tool to guide SNIP surgery. Methods In five patients diagnosed with SNIP, 10 mg of bevacizumab-800CW was intravenously administered 3 days prior to surgery. Fluorescence molecular imaging was performed in vivo during surgery and ex vivo during the processing of the surgical specimen. Fluorescence signals were correlated with final histopathology and VEGF-A immunohistochemistry. We introduced a fluorescence grid analysis to assess the fluorescence signal in individual tissue fragments, due to the nature of the surgical procedure (i.e., piecemeal resection) allowing the detection of small SNIP residues and location of the tracer ex vivo. Results In all patients, fluorescence signal was detected in vivo during endoscopic SNIP surgery. Using ex vivo fluorescence grid analysis, we were able to correlate bevacizumab-800CW fluorescence of individual tissue fragments with final histopathology. Fluorescence grid analysis showed substantial variability in mean fluorescence intensity (FImean), with SNIP tissue showing a median FImean of 77.54 (IQR 50.47–112.30) compared to 35.99 (IQR 21.48–57.81) in uninvolved tissue (p < 0.0001), although the diagnostic ability was limited with an area under the curve of 0.78. Conclusions A fluorescence grid analysis could serve as a valid method to evaluate fluorescence molecular imaging in piecemeal surgeries. As such, although substantial differences were observed in fluorescence intensities, VEGF-A may not be the ideal target for SNIP surgery. Trial registration NCT03925285. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05567-x.
Collapse
|
15
|
Development and Characterisation of Antibody-Based Optical Imaging Probes for Inflammatory Bowel Disease. Pharmaceuticals (Basel) 2021; 14:ph14090922. [PMID: 34577622 PMCID: PMC8468533 DOI: 10.3390/ph14090922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 01/07/2023] Open
Abstract
Monoclonal antibodies are an important addition to the medicinal treatment paradigm for IBD patients. While effective, these agents show a high degree of primary and secondary non-response, and methods to predict response are highly desired. Information on drug distribution at the target level is often lacking. Fluorescent endoscopic imaging using labelled antibody drugs may provide insight regarding drug distribution, target engagement and drug response, but these assessments require stable and functional fluorescently-conjugated probes. Infliximab, vedolizumab, adalimumab and ustekinumab were conjugated to IRDye 800CW, IRDye 680LT and ZW800-1. The resulting 12 tracer candidates were analysed and characterised on SE-HPLC, SDS-PAGE, iso-electric focussing (IEF) and ELISA in order to evaluate their feasibility as candidate clinical tracers for cGMP development. Major differences in the conjugation results could be seen for each conjugated drug. For Infliximab, 2 conjugates (800CW and 680LT) showed formation of aggregates, while conjugates of all drugs with ZW800-1 showed reduced fluorescent brightness, reduced purification yield and formation of fragments. All 6 of these candidates were considered unfeasible. From the remaining 6, ustekinumab-680LT showed reduced binding to IL23, and was therefore considered unfeasible. Out of 12 potential tracer candidates, 5 were considered feasible for further development: vedolizumab-800CW, vedolizumab-680LT, adalimumab-800CW, adalimumab-680LT and ustekinumab-800CW. Infliximab-680LT and ustekinumab-680LT failed to meet the standards for this panel, but may be rendered feasible if tracer production methods were further optimized.
Collapse
|
16
|
Pei J, Juniper G, van den Berg NS, Nisho N, Broadt T, Welch AR, Yi GS, Raymundo RC, Chirita SU, Lu G, Krishnan G, Lee YJ, Kapoor S, Zhou Q, Colevas AD, Lui NS, Poultsides GA, Li G, Zinn KR, Rosenthal EL. Safety and Stability of Antibody-Dye Conjugate in Optical Molecular Imaging. Mol Imaging Biol 2021; 23:109-116. [PMID: 32880818 PMCID: PMC9398032 DOI: 10.1007/s11307-020-01536-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/03/2020] [Accepted: 08/20/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE The development of molecularly targeted tracers is likely to improve the accuracy of diagnostic, screening, and therapeutic tools. Despite the many therapeutic antibodies that are FDA-approved with known toxicity, only a limited number of antibody-dye conjugates have been introduced to the clinic. Thorough evaluation of the safety, stability, and pharmacokinetics of antibody conjugates in the clinical setting compared with their parental components could accelerate the clinical approval of antibodies as agents for molecular imaging. Here we investigate the safety and stability of a near-infrared fluorescent dye (IRDye800CW) conjugated panitumumab, an approved therapeutic antibody, and report on the product stability, pharmacokinetics, adverse events, and QTc interval changes in patients. PROCEDURES Panitumumab-IRDye800CW was made under good manufacturing practice (GMP) conditions in a single batch on March 26, 2014, and then evaluated over 4.5 years at 0, 3, and 6 months, and then at 6-month intervals thereafter. We conducted early phase trials in head and neck, lung, pancreas, and brain cancers with panitumumab-IRDye800CW. Eighty-one patients scheduled to undergo standard-of-care surgery were infused with doses between 0.06 to 2.83 mg/kg of antibody. Patient ECGs, blood samples, and adverse events were collected over 30-day post-infusion for analysis. RESULTS Eighty-one patients underwent infusion of the study drug at a range of doses. Six patients (7.4 %) experienced an adverse event that was considered potentially related to the drug. The most common event was a prolonged QTc interval which occurred in three patients (3.7 %). Panitumumab-IRDye800CW had two OOS results at 42 and 54 months while meeting all other stability testing criteria. CONCLUSIONS Panitumumab-IRDye800CW was safe and stable to administer over a 54-month window with a low rate of adverse events (7.4 %) which is consistent with the rate associated with panitumumab alone. This data supports re-purposing therapeutic antibodies as diagnostic imaging agents with limited preclinical toxicology studies.
Collapse
Affiliation(s)
- Jacqueline Pei
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Georgina Juniper
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Nynke S van den Berg
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Naoki Nisho
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Trevor Broadt
- Biopharmaceutical Development Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Anthony R Welch
- Biological Resources Branch/DTP/DCTD, National Cancer Institute, Frederick, MD, USA
| | - Grace S Yi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Roan C Raymundo
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Stefania U Chirita
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Guolan Lu
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Giri Krishnan
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Yu-Jin Lee
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Shrey Kapoor
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Quan Zhou
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - A Dimitrios Colevas
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA
| | - Natalie S Lui
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - George A Poultsides
- Department of Surgery, Section of Surgical Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gordon Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kurt R Zinn
- Department of Radiology, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Eben L Rosenthal
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, 900 Blake Wilbur Drive, Stanford, CA, 94305, USA.
| |
Collapse
|
17
|
A Photocleavable Contrast Agent for Light-Responsive MRI. Pharmaceuticals (Basel) 2020; 13:ph13100296. [PMID: 33050049 PMCID: PMC7599822 DOI: 10.3390/ph13100296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/18/2020] [Accepted: 10/03/2020] [Indexed: 11/16/2022] Open
Abstract
Thanks to its innocuousness and high spatiotemporal resolution, light is used in several established and emerging applications in biomedicine. Among them is the modulation of magnetic resonance imaging (MRI) contrast agents’ relaxivity with the aim to increase the sensitivity, selectivity and amount of functional information obtained from this outstanding whole-body medical imaging technique. This approach requires the development of molecular contrast agents that show high relaxivity and strongly pronounced photo-responsiveness. To this end, we report here the design and synthesis of a light-activated MRI contrast agent, together with its evaluation using UV–vis spectroscopy, Fast Field Cycling (FFC) relaxometry and relaxometric measurements on clinical MRI scanners. The high relaxivity of the reported agent changes substantially upon irradiation with light, showing a 17% decrease in relaxivity at 0.23T upon irradiation with λ = 400 nm (violet) light for 60 min. On clinical MRI scanners (1.5T and 3.0T), irradiation leads to a decrease in relaxivity of 9% and 19% after 3 and 60 min, respectively. The molecular design presents an important blueprint for the development of light-activatable MRI contrast agents.
Collapse
|
18
|
Suurs FV, Qiu SQ, Yim JJ, Schröder CP, Timmer-Bosscha H, Bensen ES, Santini JT, de Vries EGE, Bogyo M, van Dam GM. Fluorescent image-guided surgery in breast cancer by intravenous application of a quenched fluorescence activity-based probe for cysteine cathepsins in a syngeneic mouse model. EJNMMI Res 2020; 10:111. [PMID: 32990883 PMCID: PMC7524956 DOI: 10.1186/s13550-020-00688-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/13/2020] [Indexed: 12/24/2022] Open
Abstract
Purpose The reoperation rate for breast-conserving surgery is as high as 15–30% due to residual tumor in the surgical cavity after surgery. In vivo tumor-targeted optical molecular imaging may serve as a red-flag technique to improve intraoperative surgical margin assessment and to reduce reoperation rates. Cysteine cathepsins are overexpressed in most solid tumor types, including breast cancer. We developed a cathepsin-targeted, quenched fluorescent activity-based probe, VGT-309, and evaluated whether it could be used for tumor detection and image-guided surgery in syngeneic tumor-bearing mice. Methods Binding specificity of the developed probe was evaluated in vitro. Next, fluorescent imaging in BALB/c mice bearing a murine breast tumor was performed at different time points after VGT-309 administration. Biodistribution of VGT-309 after 24 h in tumor-bearing mice was compared to control mice. Image-guided surgery was performed at multiple time points tumors with different clinical fluorescent camera systems and followed by ex vivo analysis. Results The probe was specifically activated by cathepsins X, B/L, and S. Fluorescent imaging revealed an increased tumor-to-background contrast over time up to 15.1 24 h post probe injection. In addition, VGT-309 delineated tumor tissue during image-guided surgery with different optical fluorescent imaging camera systems. Conclusion These results indicate that optical fluorescent molecular imaging using the cathepsin-targeted probe, VGT-309, may improve intraoperative tumor detection, which could translate to more complete tumor resection when coupled with commercially available surgical tools and techniques.
Collapse
Affiliation(s)
- Frans V Suurs
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Si-Qi Qiu
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. .,Diagnosis and Treatment Center of Breast Diseases, Affiliated Shantou Hospital, Sun Yat-Sen University, Shantou, China.
| | - Joshua J Yim
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Carolien P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hetty Timmer-Bosscha
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Elisabeth G E de Vries
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Matthew Bogyo
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gooitzen M van Dam
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. .,Department of Nuclear Medicine and Molecular Imaging and Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
19
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
20
|
Voskuil FJ, de Jongh SJ, Hooghiemstra WTR, Linssen MD, Steinkamp PJ, de Visscher SAHJ, Schepman KP, Elias SG, Meersma GJ, Jonker PKC, Doff JJ, Jorritsma-Smit A, Nagengast WB, van der Vegt B, Robinson DJ, van Dam GM, Witjes MJH. Fluorescence-guided imaging for resection margin evaluation in head and neck cancer patients using cetuximab-800CW: A quantitative dose-escalation study. Theranostics 2020; 10:3994-4005. [PMID: 32226534 PMCID: PMC7086353 DOI: 10.7150/thno.43227] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor-positive resection margins are present in up to 23% of head and neck cancer (HNC) surgeries, as intraoperative techniques for real-time evaluation of the resection margins are lacking. In this study, we investigated the safety and potential clinical value of fluorescence-guided imaging (FGI) for resection margin evaluation in HNC patients. We determined the optimal cetuximab-800CW dose by quantification of intrinsic fluorescence values using multi-diameter single-fiber reflectance, single-fiber fluorescence (MDSFR/SFF) spectroscopy. Methods: Five cohorts of three HNC patients received cetuximab-800CW systemically: three single dose cohorts (10, 25, 50 mg) and two cohorts pre-dosed with 75 mg unlabeled cetuximab (15 or 25 mg). Fluorescence visualization and MDSFR/SFF spectroscopy quantification was performed and were correlated to histopathology. Results: There were no study-related adverse events higher than Common Terminology Criteria for Adverse Events grade-II. Quantification of intrinsic fluorescence values showed a dose-dependent increase in background fluorescence in the single dose cohorts (p<0.001, p<0.001), which remained consistently low in the pre-dosed cohorts (p=0.6808). Resection margin status was evaluated with a sensitivity of 100% (4/4 tumor-positive margins) and specificity of 91% (10/11 tumor-negative margins). Conclusion: A pre-dose of 75 mg unlabeled cetuximab followed by 15 mg cetuximab-800CW was considered the optimal dose based on safety, fluorescence visualization and quantification of intrinsic fluorescence values. We were able to use a lower dose cetuximab-800CW than previously described, while remaining a high sensitivity for tumor detection due to application of equipment optimized for IRDye800CW detection, which was validated by quantification of intrinsic fluorescence values.
Collapse
|