1
|
Naffouje SA, Goto M, Coward LU, Gorman GS, Christov K, Wang J, Green A, Shilkaitis A, Das Gupta TK, Yamada T. Nontoxic Tumor-Targeting Optical Agents for Intraoperative Breast Tumor Imaging. J Med Chem 2022; 65:7371-7379. [PMID: 35544687 DOI: 10.1021/acs.jmedchem.2c00417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Precise identification of the tumor margins during breast-conserving surgery (BCS) remains a challenge given the lack of visual discrepancy between malignant and surrounding normal tissues. Therefore, we developed a fluorescent imaging agent, ICG-p28, for intraoperative imaging guidance to better aid surgeons in achieving negative margins in BCS. Here, we determined the pharmacokinetics (PK), biodistribution, and preclinical toxicity of ICG-p28. The PK and biodistribution of ICG-p28 indicated rapid tissue uptake and localization at tumor lesions. There were no dose-related effect and no significant toxicity in any of the breast cancer and normal cell lines tested. Furthermore, ICG-p28 was evaluated in clinically relevant settings with transgenic mice that spontaneously developed invasive mammary tumors. Intraoperative imaging with ICG-p28 showed a significant reduction in the tumor recurrence rate. This simple, nontoxic, and cost-effective method can offer a new approach that enables surgeons to intraoperatively identify tumor margins and potentially improves overall outcomes by reducing recurrence rates.
Collapse
Affiliation(s)
- Samer A Naffouje
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Masahide Goto
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Lori U Coward
- McWhorter School of Pharmacy, Pharmaceutical, Social and Administrative Sciences, Samford University, Birmingham, Alabama 35229, United States
| | - Gregory S Gorman
- McWhorter School of Pharmacy, Pharmaceutical, Social and Administrative Sciences, Samford University, Birmingham, Alabama 35229, United States
| | - Konstantin Christov
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Jing Wang
- Department of Mathematics, Statistics and Computer Science, University of Illinois College of Liberal Arts and Sciences, Urbana, Illinois 60612, United States
| | - Albert Green
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Anne Shilkaitis
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Tapas K Das Gupta
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, Illinois 60612, United States.,Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Medicine and Engineering, Chicago, Illinois 60607, United States
| |
Collapse
|
2
|
Cui L, Gao Y, Yu H, Li M, Wang B, Zhou T, Hu Q. Intraoperative Parathyroid Localization with Near-Infrared Fluorescence Imaging Using Indocyanine Green during Total Parathyroidectomy for Secondary Hyperparathyroidism. Sci Rep 2017; 7:8193. [PMID: 28811539 PMCID: PMC5557759 DOI: 10.1038/s41598-017-08347-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/11/2017] [Indexed: 12/28/2022] Open
Abstract
The detection of all glands during total parathyroidectomy (TPTX) in secondary hyperparathyroidism (SHPT) patients is often difficult due to their variability in number and location. The objective of this study was to evaluate the feasibility of near-infrared fluorescence (NIRF) imaging using indocyanine green (ICG) for intraoperative parathyroid gland (PTG) localization in SHPT patients. Twenty-nine patients with SHPT were divided into two groups with or without intraoperative NIRF imaging. ICG was administered in patients undergoing intraoperative imaging, and the fluorescence of PTGs was assessed. Clinical and histopathologic variables were analyzed to determine factors associated with ICG uptake. Comparisons between NIRF and preoperative imaging, as well as differences between groups with or without NIRF imaging, were carried out to evaluate the efficacy of this technique. Most PTGs could be clearly identified, including one ectopic gland. The sensitivity of NIRF imaging is 91.1% in contrast to 81.82% for ultrasonography (US), 62.34% for 99mTc-MIBI and 85.71% for computed tomography (CT). In addition, intraoperative NIRF imaging can reduce the operation time and improve the complete resection rate compared with the group not using it. Intraoperative NIRF imaging using ICG during TPTX is technically feasible and reliable for assisting surgeons in detecting and confirming PTGs.
Collapse
Affiliation(s)
- Le Cui
- Department of Breast and Thyroid Surgery, Puai Hospital, Wuhan, 430030, China
| | - Yang Gao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Heping Yu
- Department of Breast and Thyroid Surgery, Puai Hospital, Wuhan, 430030, China
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Birong Wang
- Department of Breast and Thyroid Surgery, Puai Hospital, Wuhan, 430030, China.
| | - Tao Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Qinggang Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
3
|
Saccomano M, Dullin C, Alves F, Napp J. Preclinical evaluation of near-infrared (NIR) fluorescently labeled cetuximab as a potential tool for fluorescence-guided surgery. Int J Cancer 2016; 139:2277-89. [PMID: 27428782 DOI: 10.1002/ijc.30277] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/01/2016] [Accepted: 06/23/2016] [Indexed: 02/06/2023]
Abstract
The high rate of recurrence in patients with pancreatic ductal adenocarcinoma (PDAC) could be reduced by supporting the surgeons in discriminating healthy from diseased tissues with intraoperative fluorescence-guidance. Here, we studied the suitability of Cetuximab, a therapeutic monoclonal antibody targeting the human epidermal growth factor receptor (EGFR), near-infrared (NIR) fluorescently labeled as a new tool for fluorescence-guided surgery. Distribution and binding of systemically injected Cetuximab Alexa Fluor 647 conjugate (Cetux-Alexa-647) and the co-injected control human IgG Alexa Fluor 750 conjugate (hIgG-Alexa-750) was studied over 48 h by NIR fluorescence imaging in mice bearing human orthotopic AsPC-1 and MIA PaCa-2 PDAC tumors. Cetux-Alexa-647, but not the control hIgG-Alexa-750 fluorescence, was specifically detected in vivo in both primary pancreatic tumors with maximum fluorescence intensities at 24 h, and in metastases of AsPC-1 tumors as small as 1 mm. Lifetime analysis and NIR fluorescence microscopy of tumor sections confirmed the binding specificity of Cetux-Alexa-647 to PDAC cells. Comparable results were obtained with Cetuximab conjugated to Alexa Fluor 750 dye (Cetux-Alexa-750). Fluorescence-guided dissection, performed 24 h after injection of Cetuximab conjugated to IRDye 800CW (Cetux-800CW), enabled a real-time delineation of AsPC-1 tumor margins, and small metastases. Odyssey scans revealed that only the vital part of the tumor, but not the necrotic part was stained with Cetux-800CW. NIR fluorescently labeled Cetuximab may be a promising tool that can be applied for fluorescence-guided surgery to visualize tumor margins and metastatic sites in order to allow a precise surgical resection.
Collapse
Affiliation(s)
- Mara Saccomano
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Christian Dullin
- Institute of Interventional and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Frauke Alves
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany.,Institute of Interventional and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany.,Department of Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Joanna Napp
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany. .,Institute of Interventional and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany. .,Department of Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
4
|
Gao M, Su H, Lin G, Li S, Yu X, Qin A, Zhao Z, Zhang Z, Tang BZ. Targeted imaging of EGFR overexpressed cancer cells by brightly fluorescent nanoparticles conjugated with cetuximab. NANOSCALE 2016; 8:15027-15032. [PMID: 27468980 DOI: 10.1039/c6nr04439e] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
To improve the treatment efficiency and reduce side effects in cancer therapy, accurate diagnosis of cancer cell types at a molecular level is highly desirable. Fluorescent nanoparticles (NPs) are especially suitable for detecting molecular biomarkers of cancer with advantages of superior brightness, easy decoration and high resolution. However, the conventional organic fluorophores, conjugated polymers, and inorganic quantum dots suffer from the drawbacks of aggregation-caused quenching (ACQ), low photostability, and heavy metal toxicity, respectively, which severely restrict their applications in NPs-based fluorescence imaging. To overcome these limitations, herein, we have developed fluorescent nanoparticles based on a t-BuPITBT-TPE fluorophore derived from aggregation-induced emission (AIE)-active tetraphenylethene. Through encapsulating t-BuPITBT-TPE within biocompatible DSPE-PEG and further decorating with a monoclonal antibody cetuximab (C225), the obtained t-BuPITBT-TPE-C225 NPs can be used for targeted imaging of non-small cell lung cancer cells with an overexpressed epidermal growth factor receptor (EGFR). The specific targeting ability of t-BuPITBT-TPE-C225 NPs has been well verified by confocal microscopy and flow cytometry experiments. The t-BuPITBT-TPE-C225 NPs have shown significant advantages in terms of highly efficient red emission, good bio-compatibility, and excellent photostability. This work provides a promising method for precise diagnosis of cancer cells by antibody-functionalized fluorescent NPs with high brightness.
Collapse
Affiliation(s)
- Meng Gao
- State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China.
| | - Huifang Su
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Gengwei Lin
- State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China.
| | - Shiwu Li
- State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China.
| | - Xingsu Yu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Anjun Qin
- State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China.
| | - Zujin Zhao
- State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China.
| | - Zhenfeng Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Ben Zhong Tang
- State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China. and Department of Chemistry, The Hong Kong University of Science & Technology, Clear Water Bay, Kowloon, Hong Kong, China. and Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Hong Kong, China
| |
Collapse
|
5
|
Zhou Y, Kim YS, Milenic D, Baidoo K, Brechbiel MW. In vitro and in vivo analysis of indocyanine green-labeled panitumumab for optical imaging-a cautionary tale. Bioconjug Chem 2014; 25:1801-10. [PMID: 25243604 PMCID: PMC4198103 DOI: 10.1021/bc500312w] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/08/2014] [Indexed: 01/17/2023]
Abstract
Indocyanine green (IC-Green), the only FDA approved near-infrared (NIR) fluorophore for clinical use, is attractive to researchers for the development of targeted optical imaging agents by modification of its structure and conjugation to monoclonal antibodies (mAbs) or their fragments. IC-Green derivative, ICG-sulfo-OSu (ICG-sOSu), is frequently used for antibody conjugation. However, ICG-sOSu is amphiphilic and readily facilitates aggregation of mAbs that is not easily separable from the desired immunoconjugates. Complications originating from this behavior are frequently overlooked by researchers. This study examined detailed chemical and biological characteristics of an ICG-sOSu-labeled mAb, panitumumab, and provided a clinically applicable strategy to deliver a pure conjugation product. Size-exclusion high-performance liquid chromatography (SE-HPLC) analysis of conjugation reactions, performed at molar reaction ratios of ICG-sOSu: mAb of 5, 10, or 20, resulted in isolable desired ICG-sOSu-panitumumab conjugation product in 72%, 53%, and 19% yields, respectively, with the remainder consisting of high molecular weight aggregates (>150 kDa) 14%, 30%, and 51%, respectively. The HPLC-purified ICG-sOSu-panitumumab products were analyzed by native and SDS polyacrylamide gel electrophoresis (PAGE) followed by optical imaging. Results indicated that the interaction between ICG-sOSu and panitumumab was due to both covalent and noncovalent binding of the ICG-sOSu to the protein. Noncovalently bound dye in the ICG-sOSu-panitumumab conjugate products was removed by extraction with ethyl acetate to further purify the HPLC-isolated conjugation products. With conserved immunoreactivity, excellent target-specific uptake of the doubly purified bioconjugates was observed with minimal liver retention in athymic nude mice bearing HER1-expressing tumor xenografts. In summary, the preparation of well-defined bioconjugate products labeled with commercial ICG-sOSu dye is not a simple process and control of the conjugation reaction ratio and conditions is crucial. Furthermore, absolute purification and characterization of the products is necessitated prior to in vivo optical imaging. Use of validated and characterized dye conjugate products should facilitate the development of clinically viable and reproducible IC-Green derivative and other NIR dye mAb conjugates for optical imaging applications.
Collapse
Affiliation(s)
- Yang Zhou
- Radioimmune & Inorganic
Chemistry Section, Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, Bethesda, Maryland 20892, United
States
| | - Young-Seung Kim
- Radioimmune & Inorganic
Chemistry Section, Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, Bethesda, Maryland 20892, United
States
| | - Diane
E. Milenic
- Radioimmune & Inorganic
Chemistry Section, Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, Bethesda, Maryland 20892, United
States
| | - Kwamena
E. Baidoo
- Radioimmune & Inorganic
Chemistry Section, Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, Bethesda, Maryland 20892, United
States
| | - Martin W. Brechbiel
- Radioimmune & Inorganic
Chemistry Section, Radiation Oncology Branch, National Cancer Institute, 10 Center Drive, Bethesda, Maryland 20892, United
States
| |
Collapse
|
6
|
Successful in vivo tumor visualization using fluorescence laparoscopy in a mouse model of disseminated alveolar rhabdomyosarcoma. Surg Endosc 2014; 29:1105-14. [PMID: 25149634 DOI: 10.1007/s00464-014-3770-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 07/25/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND Surgery for rhabdomyosarcoma is challenging due to a lack of clear delineation between tumor and surrounding tissue. Mutilating surgery can be necessary in difficult tumor localizations. Therefore, novel diagnostic and therapeutic modalities are required. The aim of this study was to evaluate the in vivo tumor detection of RMS using fluorescence laparoscopy and to analyze the efficacy of hypericin-induced photodynamic therapy in a mouse model. METHODS Seventeen NOD/LtSz-scid IL2Rγnull-mice were divided into four groups. In group 1, mCherry-expressing tumor cells and in group 2-4 non-transfected tumor cells were xenotransplanted. Three weeks later, one fluorochrome per group (ICG, ICG-cetuximab, hypericin) was injected. Fluorescence laparoscopy was carried out and tumors were resected using fluorescence guidance. In the hypericin group, photodynamic therapy was performed using blue light and apoptosis was evaluated by TUNEL test. RESULTS A clear discrimination between healthy and tumor tissue was feasible by fluorescending properties with mCherry expressing tumor cells and after injection of hypericin. No fluorescence was detected in mice injected with ICG and ICG-labeled cetuximab. Hypericin photodynamic therapy induced apoptosis of tumor cells after exposure to blue light. CONCLUSIONS Intraoperative photodynamic diagnosis was feasible using mCherry-transfected tumor cells or hypericin. Additionally, intraoperative photodynamic therapy was possible and effective.
Collapse
|
7
|
Scheuer W, van Dam GM, Dobosz M, Schwaiger M, Ntziachristos V. Drug-based optical agents: infiltrating clinics at lower risk. Sci Transl Med 2012; 4:134ps11. [PMID: 22593172 DOI: 10.1126/scitranslmed.3003572] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Fluorescent agents with specificity to cellular and subcellular moieties present promise for enhancing diagnostics and theranostics, yet challenges associated with regulatory approvals of experimental agents stifle the clinical translation. As a result, targeted fluorescent agents have remained predominantly as preclinical imaging tools. We discuss the potential of using optically labeled drugs to accelerate the clinical acceptance of optical and optoacoustic agents, in analogy to nuclear medicine approaches. This strategy, corroborated with microdosing studies, outlines a promising approach for overcoming bottlenecks and advancing photonic clinical imaging.
Collapse
Affiliation(s)
- Werner Scheuer
- Oncology Research, Roche Diagnostics GmbH, Penzberg 82377, Germany
| | | | | | | | | |
Collapse
|
8
|
Heath CH, Deep NL, Sweeny L, Zinn KR, Rosenthal EL. Use of panitumumab-IRDye800 to image microscopic head and neck cancer in an orthotopic surgical model. Ann Surg Oncol 2012; 19:3879-87. [PMID: 22669455 PMCID: PMC3595117 DOI: 10.1245/s10434-012-2435-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Indexed: 01/02/2023]
Abstract
BACKGROUND Fluorescence imaging hardware (SPY) has recently been developed for intraoperative assessment of blood flow via detection of probes emitting in the near-infrared (NIR) spectrum. This study sought to determine if this imaging system was capable of detecting micrometastatic head and neck squamous cell carcinoma (HNSCC) in preclinical models. METHODS A NIR fluorescent probe (IRDye800CW) was covalently linked to a monoclonal antibody targeting epidermal growth factor receptor (EGFR; panitumumab) or nonspecific IgG. HNSCC flank (SCC-1) and orthotopic (FADU and OSC19) xenografts were imaged 48-96 h after systemic injection of labeled panitumumab or IgG. The primary tumor and regional lymph nodes were dissected using fluorescence guidance with the SPY system and grossly assessed with a charge-coupled NIR system (Pearl). Histologic slides were also imaged with a NIR charged-coupled device (Odyssey) and fluorescence intensity was correlated with pathologic confirmation of disease. RESULTS Orthotopic tongue tumors were clearly delineated from normal tissue with tumor-to-background ratios of 2.9 (Pearl) and 2.3 (SPY). Disease detection was significantly improved with panitumumab-IRDye compared to IgG-IRDye800 (P < 0.05). Tissue biopsy samples (average size 3.7 mm) positive for fluorescence were confirmed for pathologic disease by histology and immunohistochemistry (n = 25 of 25). Biopsy samples of nonfluorescent tissue were proven to be negative for malignancy (n = 28 of 28). The SPY was able to detect regional lymph node metastasis (<1.0 mm) and microscopic areas of disease. Standard histological assessment in both frozen and paraffin-embedded histologic specimens was augmented using the Odyssey. CONCLUSIONS Panitumumab-IRDye800 may have clinical utility in detection and removal of microscopic HNSCC using existing intraoperative optical imaging hardware and may augment analysis of frozen and permanent pathology.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Carcinoma, Squamous Cell/diagnosis
- Carcinoma, Squamous Cell/surgery
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/immunology
- Head and Neck Neoplasms/diagnosis
- Head and Neck Neoplasms/surgery
- Humans
- Immunoenzyme Techniques
- Indoles
- Lymphatic Metastasis
- Mice
- Mice, SCID
- Microscopy, Fluorescence
- Models, Anatomic
- Optical Imaging
- Panitumumab
- Spectroscopy, Near-Infrared
- Surgery, Computer-Assisted
- Transplantation, Heterologous
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- C. Hope Heath
- Department of Surgery, University of Alabama at Birmingham,
Birmingham, AL 35294
| | - Nicholas L. Deep
- Department of Surgery, University of Alabama at Birmingham,
Birmingham, AL 35294
| | - Larissa Sweeny
- Department of Surgery, University of Alabama at Birmingham,
Birmingham, AL 35294
| | - Kurt R Zinn
- Radiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Eben L. Rosenthal
- Department of Surgery, University of Alabama at Birmingham,
Birmingham, AL 35294
| |
Collapse
|
9
|
Cohen R, Stammes MA, de Roos IH, Stigter-van Walsum M, Visser GW, van Dongen GA. Inert coupling of IRDye800CW to monoclonal antibodies for clinical optical imaging of tumor targets. EJNMMI Res 2011; 1:31. [PMID: 22214225 PMCID: PMC3250998 DOI: 10.1186/2191-219x-1-31] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 12/01/2011] [Indexed: 11/10/2022] Open
Abstract
Background Photoimmunodetection, in which monoclonal antibodies [mAbs] are labeled with fluorescent dyes, might have clinical potential for early detection and characterization of cancer. For this purpose, the dye should be coupled in an inert way to mAb. In this study, different equivalents of IRDye800CW, a near-infrared fluorescent dye, were coupled to 89Zr-labeled cetuximab and bevacizumab, and conjugates were evaluated in biodistribution studies. Radiolabeled mAbs were used to allow accurate quantification for assessment of the number of dye groups that can be coupled to mAbs without affecting their biological properties. Methods 89Zr-cetuximab and 89Zr-bevacizumab, containing 0.5 89Zr-desferal group per mAb molecule, were incubated with 1 to 10 eq IRDye800CW at pH 8.5 for 2 h at 35°C, and 89Zr-mAb-IRDye800CW conjugates were purified by a PD10 column using 0.9% NaCl as eluent. HPLC analysis at 780 nm was used to assess conjugation efficiency. In vitro stability measurements were performed in storage buffer (0.9% NaCl or PBS) at 4°C and 37°C and human serum at 37°C. 89Zr-mAb-IRDye800CW conjugates and 89Zr-mAb conjugates (as reference) were administered to nude mice bearing A431 (cetuximab) or FaDu (bevacizumab) xenografts, and biodistribution was assessed at 24 to 72 h after injection. Results Conjugation efficiency of IRDye800CW to 89Zr-mAbs was approximately 50%; on an average, 0.5 to 5 eq IRDye800CW was conjugated. All conjugates showed optimal immunoreactivity and were > 95% stable in storage buffer at 4°C and 37°C and human serum at 37°C for at least 96 h. In biodistribution studies with 89Zr-cetuximab-IRDye800CW, enhanced blood clearance with concomitant decreased tumor uptake and increased liver uptake was observed at 24 to 72 h post-injection when 2 or more eq of dye had been coupled to mAb. No significant alteration of biodistribution was observed 24 to 48 h after injection when 1 eq of dye had been coupled. 89Zr-bevacizumab-IRDye800CW showed a similar tendency, with an impaired biodistribution when 2 eq of dye had been coupled to mAb. Conclusion Usage of 89Zr-mAbs allows accurate quantification of the biodistribution of mAbs labeled with different equivalents of IRDye800CW. Alteration of biodistribution was observed when more than 1 eq of IRDye800CW was coupled to mAbs.
Collapse
Affiliation(s)
- Ruth Cohen
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center, De Boelelaan 1117, P,O, Box 7057, Amsterdam, 1007 MB, The Netherlands.
| | | | | | | | | | | |
Collapse
|
10
|
Marshall MV, Draney D, Sevick-Muraca EM, Olive DM. Single-dose intravenous toxicity study of IRDye 800CW in Sprague-Dawley rats. Mol Imaging Biol 2011; 12:583-94. [PMID: 20376568 PMCID: PMC2978892 DOI: 10.1007/s11307-010-0317-x] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Objective Fluorophore-labeled contrast imaging agents are moving toward clinical use for a number of applications. The near-infrared dye IRDye 800CW is frequently used in its N-hydroxysuccinamide (NHS) ester form for labeling these agents. Following conjugation or breakdown of a labeled ligand, excess NHS ester is converted to the carboxylate form. To prepare for clinical use as a near-infrared fluorophore, a toxicity study was conducted on IRDye 800CW carboxylate. Methods Male and female Sprague–Dawley rats were given a single intravenous or intradermal administration of IRDye 800CW carboxylate; Indocyanine Green was used as a comparative control. Animals were injected with varying doses of the test and control articles and observed for up to 14 days. Clinical chemistry, hematological, and pharmacokinetic analyses were performed on subgroups of animals. Organs were analyzed for content of the test article. Tissues were analyzed microscopically for pathological changes. Results Based on hematologic, clinical chemistry, and histopathologic evaluation, single administration of IRDye 800CW carboxylate intravenously at dose levels of 1, 5, and 20 mg/kg or 20 mg/kg intradermally produced no pathological evidence of toxicity. Conclusion A dose of 20 mg/kg was identified as the no observed adverse effect level following IV or ID routes of administration of IRDye 800CW. Electronic supplementary material The online version of this article (doi:10.1007/s11307-010-0317-x) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Keereweer S, Sterenborg HJCM, Kerrebijn JDF, Van Driel PBAA, Baatenburg de Jong RJ, Löwik CWGM. Image-guided surgery in head and neck cancer: current practice and future directions of optical imaging. Head Neck 2011; 34:120-6. [PMID: 21284051 DOI: 10.1002/hed.21625] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 08/12/2010] [Indexed: 11/08/2022] Open
Abstract
A key aspect for the postoperative prognosis of patients with head and neck cancer is complete tumor resection. In current practice, the intraoperative assessment of the tumor-free margin is dependent on visual appearance and palpation of the tumor. Optical imaging has the potential of traversing the gap between radiology and surgery by providing real-time visualization of the tumor, thereby allowing for image-guided surgery. The use of the near-infrared light spectrum offers 2 essential advantages: increased tissue penetration of light and an increased signal-to-background ratio of contrast agents. In this review, the current practice and limitations of image-guided surgery by optical imaging using intrinsic fluorescence or contrast agents are described. Furthermore, we provide an overview of the various molecular contrast agents targeting specific hallmarks of cancer that have been used in other fields of oncologic surgery, and we describe perspectives on its future use in head and neck cancer surgery.
Collapse
Affiliation(s)
- S Keereweer
- Department of Otorhinolaryngology-Head and Neck Surgery, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
12
|
Portnoy E, Lecht S, Lazarovici P, Danino D, Magdassi S. Cetuximab-labeled liposomes containing near-infrared probe for in vivo imaging. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:480-8. [PMID: 21272665 DOI: 10.1016/j.nano.2011.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 12/23/2010] [Accepted: 01/10/2011] [Indexed: 02/08/2023]
Abstract
UNLABELLED A new liposome-based near-infrared probe that combines both imaging and targeting abilities was developed for application in medical imaging. The near-infrared fluorescent molecule indocyanine green (ICG), and the cetuximab monoclonal antibody for epidermal growth factor receptor (EGFR) were attached to liposomes by passive adsorption. It was found that ICG molecules adsorbed to the liposomes are more fluorescent than free ICG and have a larger quantum yield. Cetuximab-adsorbed fluorescent liposomes preserved EGFR recognition, as is evident from internalization and selective binding to A431 colon carcinoma cells overexpressing EGFR. The binding of cetuximab-targeted fluorescent liposomes to A431 compared with IEC-6 cells (normal enterocytes expressing physiological EGFR levels) was greater by a factor of 3.5, ensuring imaging abilities with available fluorescent equipment. Due to relatively high quantum yield and specific tumor cell-recognizing ability, this technology deserves further in vivo evaluation for imaging and diagnostic purposes. FROM THE CLINICAL EDITOR A new liposome-based near-infrared probe combining both imaging and targeting abilities is reported. Due to relatively high quantum yield and EGFR-expressing tumor cell specificity, this technology deserves further in vivo evaluation for imaging and diagnostic purposes.
Collapse
Affiliation(s)
- Emma Portnoy
- Casali Institute of Applied Chemistry, Institute of Chemistry and Center for Nanoscience and Nanotechnology, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
13
|
Villaraza AJL, Milenic DE, Brechbiel MW. Improved speciation characteristics of PEGylated indocyanine green-labeled Panitumumab: revisiting the solution and spectroscopic properties of a near-infrared emitting anti-HER1 antibody for optical imaging of cancer. Bioconjug Chem 2010; 21:2305-12. [PMID: 21073171 DOI: 10.1021/bc100336b] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A water-soluble amine-reactive PEGylated analogue of near-infrared emitting dye indocyanine green (5) was synthesized and used to label the anti-HER1 antibody panitumumab (Vectibix) at various equivalents. These conjugates were compared with non-PEGylated analogue conjugate products and the solution speciation analyzed with UV-vis spectrophotometry, size exclusion HPLC, and SDS-PAGE. PEGylation of the bioconjugates was successful in preventing aggregation in solution, a phenomenon observed with the non-PEGylated bioconjugates presumably due to the hydrophobicity of indocyanine green. Competitive radioimmunoassay demonstrated that the targeting moiety of the PEGylated bioconjugates was conserved. Fluorescence microscopy also demonstrated membrane binding of the bioconjugate to HER1-expressing A431 cells. Hence, these bioconjugates are suitable candidates for the in vivo optical imaging of HER1-expressing tumors.
Collapse
Affiliation(s)
- Aaron Joseph L Villaraza
- Radioimmune and Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1002, USA
| | | | | |
Collapse
|
14
|
A parallel excitation based fluorescence molecular tomography system for whole-body simultaneous imaging of small animals. Ann Biomed Eng 2010; 38:3440-8. [PMID: 20544284 DOI: 10.1007/s10439-010-0093-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 05/30/2010] [Indexed: 10/19/2022]
Abstract
Challenges remain in imaging complete dynamic physiological processes in vivo through the whole small animal body using fluorescence molecular tomography (FMT). In this article, a novel non-contact full-angle FMT system that enables whole-body simultaneous imaging of small animals is presented. The whole-body simultaneous imaging ability is achieved by employing a line-shaped parallel excitation source, which can provide extended spatial sampling dataset to reconstruct multiple fluorescent targets distributed in whole animal body during one full-angle FMT imaging process. The key performances of this system were evaluated by a series of experiments. Quantitation linearity for over two orders of magnitude of fluorescence markers concentration was demonstrated, and an accessible simultaneous imaging domain of 4.0 x 1.5 cm² could be achieved utilizing the parallel excitation pattern. Moreover, the in vivo imaging feasibility and performance were validated by localizing two fluorescent targets implanted at different positions of a nude mouse. The results suggest that compared with conventional single point excitation FMT system, the proposed system can achieve a whole-body simultaneous imaging domain and impart the ability to image complete dynamic physiological processes in vivo.
Collapse
|
15
|
Rapid optical imaging of EGF receptor expression with a single-chain antibody SNAP-tag fusion protein. Eur J Nucl Med Mol Imaging 2010; 37:1926-34. [PMID: 20449589 DOI: 10.1007/s00259-010-1482-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 04/13/2010] [Indexed: 12/11/2022]
Abstract
PURPOSE The epidermal growth factor receptor (EGFR) is overexpressed in several types of cancer and its inhibition can effectively inhibit tumour progression. The purpose of this study was to design an EGFR-specific imaging probe that combines efficient tumour targeting with rapid systemic clearance to facilitate non-invasive assessment of EGFR expression. METHODS Genetic fusion of a single-chain antibody fragment with the SNAP-tag produced a 48-kDa antibody derivative that can be covalently and site-specifically labelled with substrates containing 0 (6)-benzylguanine. The EGFR-specific single-chain variable fragment (scFv) fusion protein 425(scFv)SNAP was labelled with the near infrared (NIR) dye BG-747, and its accumulation, specificity and kinetics were monitored using NIR fluorescence imaging in a subcutaneous pancreatic carcinoma xenograft model. RESULTS The 425(scFv)SNAP fusion protein accumulates rapidly and specifically at the tumour site. Its small size allows efficient renal clearance and a high tumour to background ratio (TBR) of 33.2 +/- 6.3 (n = 4) 10 h after injection. Binding of the labelled antibody was efficiently competed with a 20-fold excess of unlabelled probe, resulting in an average TBR of 6 +/- 1.35 (n = 4), which is similar to that obtained with a non-tumour-specific probe (5.44 +/- 1.92, n = 4). When compared with a full-length antibody against EGFR (cetuximab), 425(scFv)SNAP-747 showed significantly higher TBRs and complete clearance 72 h post-injection. CONCLUSION The 425(scFv)SNAP fusion protein combines rapid and specific targeting of EGFR-positive tumours with a versatile and robust labelling technique that facilitates the attachment of fluorophores for use in optical imaging. The same approach could be used to couple a chelating agent for use in nuclear imaging.
Collapse
|
16
|
Ogawa M, Regino CAS, Seidel J, Green MV, Xi W, Williams M, Kosaka N, Choyke PL, Kobayashi H. Dual-modality molecular imaging using antibodies labeled with activatable fluorescence and a radionuclide for specific and quantitative targeted cancer detection. Bioconjug Chem 2010; 20:2177-84. [PMID: 19919110 DOI: 10.1021/bc900362k] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Multimodality molecular imaging should have potential for compensating the disadvantages and enhancing the advantages of each modality. Nuclear imaging is superior to optical imaging in whole body imaging and in quantification due to good tissue penetration of gamma rays. However, target specificity can be compromised by high background signal due to the always signal ON feature of nuclear probes. In contrast, optical imaging can be superior in target-specific imaging by employing target-specific signal activation systems, although it is not quantitative because of signal attenuation. In this study, to take advantage of the mutual cooperation of each modality, multimodality imaging was performed by a combination of quantitative radiolabeled probe and an activatable optical probe. The monoclonal antibodies, panitumumab (anti-HER1) and trastuzumab (anti-HER2), were labeled with 111In and ICG and tested in both HER1 and HER2 tumor bearing mice by the cocktail injection of radiolabeled and optical probes and by the single injection of a dual-labeled probe. The optical and nuclear images were obtained over 6 days after the conjugates injection. The fluorescence activation properties of ICG labeled antibodies were also investigated by in vitro microscopy. In vitro microscopy demonstrated that there was no fluorescence signal with either panitumumab-ICG or trastuzumab-ICG, when the probes were bound to cell surface antigens but were not yet internalized. After the conjugates were internalized into the cells, both conjugates showed bright fluorescence signal only in the target cells. These results show that both conjugates work as activatable probes. In in vivo multimodality imaging by injection of a cocktail of radio-optical probes, only the target specific tumor was visualized by optical imaging. Meanwhile, the biodistribution profile of the injected antibody was provided by nuclear imaging. Similar results were obtained with radio and optical dual-labeled probes, and it is confirmed that pharmacokinetic properties did not affect the results above. Here, we could characterize the molecular targets by activatable optical probes and visualize the delivery of targeting molecules quantitatively by radioactive probes. Multimodality molecular imaging combining activatable optical and radioactive probes has great potential for simultaneous visualization, characterization, and measurement of biological processes.
Collapse
Affiliation(s)
- Mikako Ogawa
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|