1
|
Jeon OH, Kim K, Kim CG, Choi BH, Lee JH, Kim BM, Kim HK. Novel locally nebulized indocyanine green for simultaneous identification of tumor margin and intersegmental plane. Int J Surg 2024; 110:4708-4715. [PMID: 38752517 PMCID: PMC11325942 DOI: 10.1097/js9.0000000000001581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/25/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Segmentectomy, recommended for early-stage lung cancer or compromised lung function, demands precise tumor detection and intersegmental plane identification. While indocyanine green (ICG) commonly aids in these aspects using near-infrared imaging, its separate administrations through different routes and times can lead to complications and patient anxiety. This study aims to develop a lung-specific delivery method by nebulizing low-dose ICG to targeted lung segments, allowing simultaneous detection of lung tumors and intersegmental planes across diverse animal models. METHODS To optimizing the dose of ICG for lung tumor and interlobar fissure detection, different doses of ICG (0.25, 0.1, and 0.05 mg/kg) were nebulized to rabbit lung tumor models. The distribution of locally nebulized ICG in targeted segments was studied to evaluate the feasibility of detecting lung tumor and intersegmental planes in canine lung pseudotumor models. RESULTS Near-infrared fluorescence imaging demonstrated clear visualization of lung tumor margin and interlobar fissure using local nebulization of 0.1 mg/kg ICG for only 4 min during surgery in the rabbit models. In the canine model, the local nebulization of 0.05 mg/kg of ICG into the target segment enabled clear visualization of pseudotumor and intersegmental planes for 30 min. CONCLUSIONS This innovative approach achieves a reduction in ICG dose and prolonged the visualization time of the intersegmental plane and effectively eliminates the need for the hurried marking of tumors and intersegmental planes. The authors anticipate that lung-specific delivery of ICG will prove valuable for image-guided limited resection of lung tumors in clinical practice.
Collapse
Affiliation(s)
- Ok Hwa Jeon
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University
- Department of Biomedical Sciences, College of Medicine, Korea University
- Image Guided Precision Cancer Surgery Institute, Seoul, Korea
| | - Kyungsu Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University
- Department of Biomedical Sciences, College of Medicine, Korea University
- Image Guided Precision Cancer Surgery Institute, Seoul, Korea
| | - Chang Geun Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University
- Department of Biomedical Sciences, College of Medicine, Korea University
- Image Guided Precision Cancer Surgery Institute, Seoul, Korea
| | - Byeong Hyeon Choi
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University
- Image Guided Precision Cancer Surgery Institute, Seoul, Korea
| | - Jun Hee Lee
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University
- Image Guided Precision Cancer Surgery Institute, Seoul, Korea
| | - Beop-Min Kim
- Department of Bio-Convergence, Korea University
- Image Guided Precision Cancer Surgery Institute, Seoul, Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University
- Department of Biomedical Sciences, College of Medicine, Korea University
- Image Guided Precision Cancer Surgery Institute, Seoul, Korea
| |
Collapse
|
2
|
Feng Q, Wang SA, Ning B, Xie J, Ding J, Liu S, Ai S, Li F, Wang X, Guan W. Evaluation of the tumor-targeting specific imaging and killing effect of a CEA-targeting nanoparticle in colorectal cancer. Biochem Biophys Res Commun 2024; 719:150084. [PMID: 38733742 DOI: 10.1016/j.bbrc.2024.150084] [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] [Received: 03/07/2024] [Revised: 04/23/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024]
Abstract
INTRODUCTION Colorectal cancer (CRC) is a prevalent digestive malignancy with significant global mortality and morbidity rates. Improving diagnostic capabilities for CRC and investigating novel therapeutic approaches are pressing clinical imperatives. Additionally, carcinoembryonic antigen (CEA) has emerged as a highly promising candidate for both colorectal tumor imaging and treatment. METHODS A novel active CEA-targeting nanoparticle, CEA(Ab)-MSNs-ICG-Pt, was designed and synthesized, which served as a tumor-specific fluorescence agent to help in CRC near-infrared (NIR) fluorescence imaging. In cell studies, CEA(Ab)-MSNs-ICG-Pt exhibited specific targeting to RKO cells through specific antibody-antigen binding of CEA, resulting in distribution both within and around these cells. The tumor-targeting-specific imaging capabilities of the nanoparticle were determined through in vivo fluorescence imaging experiments. Furthermore, the efficacy of the nanoparticle in delivering chemotherapeutics and its killing effect were evaluated both in vitro and in vivo. RESULTS The CEA(Ab)-MSNs-ICG-Pt nanoparticle, designed as a novel targeting agent for carcinoembryonic antigen (CEA), exhibited dual functionality as a targeting fluorescent agent. This CEA-targeting nanoparticle showed exceptional efficacy in eradicating CRC cells in comparison to individual treatment modalities. Furthermore, it exhibits exceptional biosafety and biocompatibility properties. CEA(Ab)-MSNs-ICG-Pt exhibits significant promise due to its ability to selectively target tumors through NIR fluorescence imaging and effectively eradicate CRC cells with minimal adverse effects in both laboratory and in vivo environments. CONCLUSION The favorable characteristics of CEA(Ab)-MSNs-ICG-Pt offer opportunities for its application in chemotherapeutic interventions, tumor-specific NIR fluorescence imaging, and fluorescence-guided surgical procedures.
Collapse
Affiliation(s)
- Qingzhao Feng
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, 210008, China; Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Shu-An Wang
- Department of Clinic Nutrition, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Beibei Ning
- Department of Pediatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China
| | - Jixian Xie
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing, Jiangsu, 211816, China
| | - Jie Ding
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Song Liu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Shichao Ai
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Fuchao Li
- Department of Gerontology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008,China
| | - Xuerui Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, Nanjing, Jiangsu, 211816, China.
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, 210008, China; Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
3
|
Kim K, Jeon OH, Choi BH, Rho J, Lee JH, Eo JS, Kim BM, Kim HK. Simultaneous Visualization of Lung Tumor and Intersegmental Plane during Pulmonary Segmentectomy by Intravenous Injection of Indocyanine Green. Cancers (Basel) 2024; 16:1439. [PMID: 38611116 PMCID: PMC11010939 DOI: 10.3390/cancers16071439] [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: 02/27/2024] [Revised: 03/22/2024] [Accepted: 04/06/2024] [Indexed: 04/14/2024] Open
Abstract
Segmentectomy is a targeted surgical approach tailored for patients with compromised health and early-stage lung cancer. The key to successful segmentectomy lies in precisely identifying the tumor and intersegmental planes to ensure adequate resection margins. In this study, we aimed to enhance this process by simultaneously visualizing the tumor and intersegmental planes through the intravenous injection of indocyanine green (ICG) at different time points and doses. Lung tumors were detected by intravenous injection of ICG at a dose of 2 mg/kg 12 h before surgery in a rabbit model. Following the dissection of the pulmonary artery, vein, and bronchi of the target segment, 0.6 mg/kg of ICG was injected intravenously to detect the intersegmental plan. Fluorescent images of the lung tumors and segments were acquired, and the fluorescent signal was quantified using the signal-to-background ratio (SBR). Finally, a pilot study of this method was conducted in three patients with lung cancer. In a preclinical study, the SBR of the tumor (4.4 ± 0.1) and nontargeted segments (10.5 ± 0.8) were significantly higher than that of the targeted segment (1.6 ± 0.2) (targeted segment vs. nontarget segment, p < 0.0001; target segment vs. tumor, p < 0.01). Consistent with preclinical results, lung tumors and the intersegmental plane were successfully detected in patients with lung cancer. Consequently, adequate resection margins were identified during the surgery, and segmentectomy was successfully performed in patients with lung cancer. This study is the first to use intravenous ICG injections at different time points and doses to simultaneously detect lung cancer and intersegmental planes, thereby achieving segmentectomy for lung cancer.
Collapse
Affiliation(s)
- Kyungsu Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea; (K.K.); (O.H.J.); (B.H.C.); (J.R.); (J.H.L.)
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ok Hwa Jeon
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea; (K.K.); (O.H.J.); (B.H.C.); (J.R.); (J.H.L.)
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Byeong Hyeon Choi
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea; (K.K.); (O.H.J.); (B.H.C.); (J.R.); (J.H.L.)
| | - Jiyun Rho
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea; (K.K.); (O.H.J.); (B.H.C.); (J.R.); (J.H.L.)
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jun Hee Lee
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea; (K.K.); (O.H.J.); (B.H.C.); (J.R.); (J.H.L.)
| | - Jae Seon Eo
- Department of Nuclear Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea;
| | - Beop-Min Kim
- Department of Biomedical Engineering, Korea University College of Health Science, Seoul 02841, Republic of Korea;
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul 02841, Republic of Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea; (K.K.); (O.H.J.); (B.H.C.); (J.R.); (J.H.L.)
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
4
|
Mi J, Li C, Yang F, Shi X, Zhang Z, Guo L, Jiang G, Li Y, Wang J, Yang F, Hu Z, Zhou J. Comparative Study of Indocyanine Green Fluorescence Imaging in Lung Cancer with Near-Infrared-I/II Windows. Ann Surg Oncol 2024; 31:2451-2460. [PMID: 38063990 DOI: 10.1245/s10434-023-14677-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/12/2023] [Indexed: 03/06/2024]
Abstract
BACKGROUND We compare the application of intravenous indocyanine green (ICG) fluorescence imaging in lung cancer with near-infrared-I (NIR-I) and near-infrared-II (NIR-II) windows. METHODS From March to December 2022, we enrolled patients who received an intravenous injection of ICG (5 mg/kg) 1 day before the planned lung cancer surgery. The lung cancer nodules were imaged by NIR-I/II fluorescence imaging systems, and the tumor-to-normal-tissue ratio (TNR) was calculated. In addition, the fluorescence intensity and signal-to-background ratio (SBR) of capillary glass tubes containing ICG covered with different thicknesses of lung tissue were measured by NIR-I/II fluorescence imaging systems. RESULTS In this study, 102 patients were enrolled, and the mean age was 59.9 ± 9.2 years. A total of 96 (94.1%) and 98 (96.1%) lung nodules were successfully imaged with NIR-I and NIR-II fluorescence, and the TNR of NIR-II was significantly higher than that of NIR-I (3.9 ± 1.3 versus 2.4 ± 0.6, P < 0.001). In multiple linear regression, solid nodules (P < 0.001) and squamous cell carcinoma (P < 0.001) were independent predictors of a higher TNR of NIR-I/II. When capillary glass tubes were covered with lung tissue whose thickness was more than 2 mm, the fluorescence intensity and the SBR of NIR-II were significantly higher than those of NIR-I. CONCLUSIONS We verified the feasibility of NIR-II fluorescence imaging in intravenous ICG lung cancer imaging for the first time. NIR-II fluorescence can improve the TNR and penetration depth of lung cancer with promising clinical prospects.
Collapse
Affiliation(s)
- Jiahui Mi
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Changjian Li
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, China
| | - Feng Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Xiaojing Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Zeyu Zhang
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, China
| | - Lishuang Guo
- School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology, Beijing, China
| | - Guanchao Jiang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Jun Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China.
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.
| | - Jian Zhou
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
5
|
Li Y, Dai C, Hua Z, Xia L, Ding Y, Wang Q, Gié MLM, Bouvet M, Cai H. A human serum albumin-indocyanine green complex offers improved tumor identification in fluorescence-guided surgery. Transl Cancer Res 2024; 13:437-452. [PMID: 38410209 PMCID: PMC10894326 DOI: 10.21037/tcr-23-2338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/18/2024] [Indexed: 02/28/2024]
Abstract
Background Complete tumor removal is critical for achieving a good prognosis in patients but remains challenging for surgeons. Near-infrared fluorescence-guided surgery (NIRFGS) enables surgeons to accurately localize tumors in real time and facilitates accurate resection. Indocyanine green (ICG) has been approved by the U.S. Food and Drug Administration and the National Medical Products Administration for many years. Although the application of ICG has progressed for a variety of surgeries, there are inherent limitations to ICG, including poor water solubility and photostability, short blood half-life, and aggregation in blood, resulting in poor imaging performance. We found that mixing ICG with human serum albumin (HSA) preoperatively and then injecting it can improve the imaging performance. Methods We prepared fluorescent probes by combining ICG with HSA and identified their optimal ratio via in vitro absorption measurement and emission spectrum characterization of ICG-HSA complex with different mixing ratios and concentration gradients. Subsequently, under the optimal ratio and clinical simulated concentration, we conducted dynamic change analysis of the fluorescence spectral properties after mixing. We then compared the uptake of ICG-HSA in vitro for two different cell types and the imaging performance of different molar ratios of ICG and HSA in mouse models. Results Through in vitro absorption and emission spectrum characterization of ICG-HSA mixtures with different mixing ratios and concentration gradients, the optimal ratio of the mixture was obtained (ICG:HSA =4:5). Using this ratio, clinical simulated concentration, and mixing, we completed the dynamic change analysis of the fluorescence spectrum properties. The results verified that HSA can improve the dispersion and stability of ICG in aqueous solution, reduce the proportion of free-state ICG, and thus improve the biodistribution. Moreover, the fluorescence performance of ICG was improved. ICG-HSA and ICG uptake in MDA-MB-231 cells and imaging in vivo showed that HSA increased the enrichment of ICG in tumor compared to ICG alone (ICG-HSAfluorescence intensity =237.3±10.7 vs. ICGfluorescence intensity =127.1±10.7). Compared with ICG alone, ICG-HSA provided a clearer tumor boundary and higher tumor-to-background ratio (TBR) (ICG-HSATBRmax 3.49±0.56 vs. ICGTBRmax 1.94±0.23). Conclusions This study suggests that ICG-HSA can achieve higher tumor-to-background contrast with shorter time and can provide an overall superior imaging performance compared to ICG alone, thus exhibiting considerable potential for clinical application.
Collapse
Affiliation(s)
- Yunlong Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Chun Dai
- Department of General Surgery, The People’s Hospital of Yangzhong City, Yangzhong, China
| | - Zhaolai Hua
- Department of General Surgery, The People’s Hospital of Yangzhong City, Yangzhong, China
| | - Lin Xia
- Department of General Surgery, The People’s Hospital of Yangzhong City, Yangzhong, China
| | - Yongbin Ding
- Department of General Surgery, Pukou Branch of Jiangsu People’s Hospital, Nanjing, China
| | - Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Marie-Laure Matthey Gié
- Department of Thoracic and Endocrine Surgery, University Hospital of Geneva (HUG), Geneva, Switzerland
- Department of Surgery, Clinica Moncucco, Lugano, Switzerland
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, CA, USA
| | - Huiming Cai
- Nanjing Nuoyuan Medical Devices Co., Ltd., Nanjing, China
| |
Collapse
|
6
|
Bianconi A, Bonada M, Zeppa P, Colonna S, Tartara F, Melcarne A, Garbossa D, Cofano F. How Reliable Is Fluorescence-Guided Surgery in Low-Grade Gliomas? A Systematic Review Concerning Different Fluorophores. Cancers (Basel) 2023; 15:4130. [PMID: 37627158 PMCID: PMC10452554 DOI: 10.3390/cancers15164130] [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: 06/29/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Fluorescence-guided surgery has been increasingly used to support glioma surgery with the purpose of obtaining a maximal safe resection, in particular in high-grade gliomas, while its role is less definitely assessed in low-grade gliomas. METHODS A systematic review was conducted. 5-aminolevulinic acid, sodium fluorescein, indocyanine green and tozuleristide were taken into account. The main considered outcome was the fluorescence rate, defined as the number of patients in whom positive fluorescence was detected out of the total number of patients. Only low-grade gliomas were considered, and data were grouped according to single fluorophores. RESULTS 16 papers about 5-aminolevulinic acid, 4 about sodium fluorescein, 2 about indocyanine green and 1 about tozuleristide were included in the systematic review. Regarding 5-aminolevulinic acid, a total of 467 low-grade glioma patients were included, and fluorescence positivity was detected in 34 out of 451 Grade II tumors (7.3%); while in Grade I tumors, fluorescence positivity was detected in 9 out of 16 cases. In 16 sodium fluorescein patients, seven positive fluorescent cases were detected. As far as indocyanine is concerned, two studies accounting for six patients (three positive) were included, while for tozuleristide, a single clinical trial with eight patients (two positive) was retrieved. CONCLUSIONS The current evidence does not support the routine use of 5-aminolevulinic acid or sodium fluorescein with a standard operating microscope because of the low fluorescence rates. New molecules, including tozuleristide, and new techniques for fluorescence detection have shown promising results; however, their use still needs to be clinically validated on a large scale.
Collapse
Affiliation(s)
- Andrea Bianconi
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Marta Bonada
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Pietro Zeppa
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Stefano Colonna
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Fulvio Tartara
- Headache Science and Neurorehabilitation Center, IRCCS Mondino Foundation, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Antonio Melcarne
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Diego Garbossa
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Fabio Cofano
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
- Humanitas Gradenigo, 10100 Turin, Italy
| |
Collapse
|
7
|
Jeon OH, Choi BH, Rho J, Kim K, Lee JH, Lee J, Kim BM, Kim HK. Optimization of Indocyanine Green for Intraoperative Fluorescent Image-Guided Localization of Lung Cancer; Analysis Based on Solid Component of Lung Nodule. Cancers (Basel) 2023; 15:3643. [PMID: 37509304 PMCID: PMC10377801 DOI: 10.3390/cancers15143643] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
ICG fluorescence imaging has been used to detect lung cancer; however, there is no consensus regarding the optimization of the indocyanine green (ICG) injection method. The aim of this study was to determine the optimal dose and timing of ICG for lung cancer detection using animal models and to evaluate the feasibility of ICG fluorescence in lung cancer patients. In a preclinical study, twenty C57BL/6 mice with footpad cancer and thirty-three rabbits with VX2 lung cancer were used. These animals received an intravenous injection of ICG at 0.5, 1, 2, or 5 mg/kg, and the cancers were detected using a fluorescent imaging system after 3, 6, 12, and 24 h. In a clinical study, fifty-one patients diagnosed with lung cancer and scheduled to undergo surgery were included. Fluorescent images of lung cancer were obtained, and the fluorescent signal was quantified. Based on a preclinical study, the optimal injection method for lung cancer detection was 2 mg/kg ICG 12 h before surgery. Among the 51 patients, ICG successfully detected 37 of 39 cases with a consolidation-to-tumor (C/T) ratio of >50% (TNR: 3.3 ± 1.2), while it failed in 12 cases with a C/T ratio ≤ 50% and 2 cases with anthracosis. ICG injection at 2 mg/kg, 12 h before surgery was optimal for lung cancer detection. Lung cancers with the C/T ratio > 50% were successfully detected using ICG with a detection rate of 95%, but not with the C/T ratio ≤ 50%. Therefore, further research is needed to develop fluorescent agents targeting lung cancer.
Collapse
Affiliation(s)
- Ok Hwa Jeon
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Byeong Hyeon Choi
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jiyun Rho
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kyungsu Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jun Hee Lee
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Jinhwan Lee
- Department of Pathology, Myongji Hospital, Goyang 10475, Republic of Korea
| | - Beop-Min Kim
- Department of Biomedical Engineering, Korea University College of Health Science, Seoul 02841, Republic of Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
8
|
Kim J, Kim H, Yoon YS, Kim CW, Hong SM, Kim S, Choi D, Chun J, Hong SW, Hwang SW, Park SH, Yang DH, Ye BD, Byeon JS, Yang SK, Kim SY, Myung SJ. Investigation of artificial intelligence integrated fluorescence endoscopy image analysis with indocyanine green for interpretation of precancerous lesions in colon cancer. PLoS One 2023; 18:e0286189. [PMID: 37228164 DOI: 10.1371/journal.pone.0286189] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Indocyanine green (ICG) has been used in clinical practice for more than 40 years and its safety and preferential accumulation in tumors has been reported for various tumor types, including colon cancer. However, reports on clinical assessments of ICG-based molecular endoscopy imaging for precancerous lesions are scarce. We determined visualization ability of ICG fluorescence endoscopy in colitis-associated colon cancer using 30 lesions from an azoxymethane/dextran sulfate sodium (AOM/DSS) mouse model and 16 colon cancer patient tissue-samples. With a total of 60 images (optical, fluorescence) obtained during endoscopy observation of mouse colon cancer, we used deep learning network to predict four classes (Normal, Dysplasia, Adenoma, and Carcinoma) of colorectal cancer development. ICG could detect 100% of carcinoma, 90% of adenoma, and 57% of dysplasia, with little background signal at 30 min after injection via real-time fluorescence endoscopy. Correlation analysis with immunohistochemistry revealed a positive correlation of ICG with inducible nitric oxide synthase (iNOS; r > 0.5). Increased expression of iNOS resulted in increased levels of cellular nitric oxide in cancer cells compared to that in normal cells, which was related to the inhibition of drug efflux via the ABCB1 transporter down-regulation resulting in delayed retention of intracellular ICG. With artificial intelligence training, the accuracy of image classification into four classes using data sets, such as fluorescence, optical, and fluorescence/optical images was assessed. Fluorescence images obtained the highest accuracy (AUC of 0.8125) than optical and fluorescence/optical images (AUC of 0.75 and 0.6667, respectively). These findings highlight the clinical feasibility of ICG as a detector of precancerous lesions in real-time fluorescence endoscopy with artificial intelligence training and suggest that the mechanism of ICG retention in cancer cells is related to intracellular nitric oxide concentration.
Collapse
Affiliation(s)
- Jinhyeon Kim
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hajung Kim
- Convergence Medicine Research Center, Asan Medical Center, Seoul, Republic of Korea
| | - Yong Sik Yoon
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chan Wook Kim
- Department of Colon and Rectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Mo Hong
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sungjee Kim
- Department of Chemistry and School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science & Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Doowon Choi
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science & Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Jihyun Chun
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung Wook Hong
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Wook Hwang
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Hyoung Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Hoon Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byong Duk Ye
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Sik Byeon
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sun Young Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Jae Myung
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Edis Biotech, Songpa-gu, Seoul, Republic of Korea
| |
Collapse
|
9
|
He S, Zhong A, Lei J, Deng Z, Zhu X, Wei R, Huang H, Chen Z, Cai L, Xie Y. Application of Indocyanine Green Fluorescence Imaging in Assisting Biopsy of Musculoskeletal Tumors. Cancers (Basel) 2023; 15:cancers15082402. [PMID: 37190330 DOI: 10.3390/cancers15082402] [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: 03/01/2023] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
(1) Background: Biopsies are the gold standard for the diagnosis of musculoskeletal tumors. In this study, we aimed to explore whether indocyanine green near-infrared fluorescence imaging can assist in the biopsy of bone and soft tissue tumors and improve the success rate of biopsy. (2) Method: We recruited patients with clinically considered bone and soft tissue tumors and planned biopsies. In the test group, indocyanine green (0.3 mg/kg) was injected. After identifying the lesion, a near-infrared fluorescence camera system was used to verify the ex vivo specimens of the biopsy in real time. If the biopsy specimens were not developed, we assumed that we failed to acquire lesions, so the needle track and needle position were adjusted for the supplementary biopsy, and then real-time imaging was performed again. Finally, we conducted a pathological examination. In the control group, normal biopsy was performed. (3) Results: The total diagnosis rate of musculoskeletal tumors in the test group was 94.92% (56/59) and that in the control group was 82.36% (42/51). In the test group, 14 cases were not developed, as seen from real-time fluorescence in the core biopsy, and then underwent the supplementary biopsy after changing the puncture direction and the location of the needle channel immediately, of which 7 cases showed new fluorescence. (4) Conclusions: Using the near-infrared fluorescence real-time development technique to assist the biopsy of musculoskeletal tumors may improve the accuracy of core biopsy and help to avoid missed diagnoses, especially for some selected tumors.
Collapse
Affiliation(s)
- Siyuan He
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ang Zhong
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jun Lei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhouming Deng
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiaobin Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Renxiong Wei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Huayi Huang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zhenyi Chen
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuanlong Xie
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
10
|
Hirano R, Okamoto K, Shinke M, Sato M, Watanabe S, Watanabe H, Kondoh G, Kadonosono T, Kizaka-Kondoh S. Tissue-resident macrophages are major tumor-associated macrophage resources, contributing to early TNBC development, recurrence, and metastases. Commun Biol 2023; 6:144. [PMID: 36737474 PMCID: PMC9898263 DOI: 10.1038/s42003-023-04525-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and highly heterogenous disease with no well-defined therapeutic targets. Treatment options are thus limited and mortality is significantly higher compared with other breast cancer subtypes. Mammary gland tissue-resident macrophages (MGTRMs) are found to be the most abundant stromal cells in early TNBC before angiogenesis. We therefore aimed to explore novel therapeutic approaches for TNBC by focusing on MGTRMs. Local depletion of MGTRMs in mammary gland fat pads the day before TNBC cell transplantation significantly reduced tumor growth and tumor-associated macrophage (TAM) infiltration in mice. Furthermore, local depletion of MGTRMs at the site of TNBC resection markedly reduced recurrence and distant metastases, and improved chemotherapy outcomes. This study demonstrates that MGTRMs are a major TAM resource and play pivotal roles in the growth and malignant progression of TNBC. The results highlight a possible novel anti-cancer approach targeting tissue-resident macrophages.
Collapse
Affiliation(s)
- Ryuichiro Hirano
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Koki Okamoto
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Miyu Shinke
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Marika Sato
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Shigeaki Watanabe
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Hitomi Watanabe
- grid.258799.80000 0004 0372 2033Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto, 606-8507 Japan
| | - Gen Kondoh
- grid.258799.80000 0004 0372 2033Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto, 606-8507 Japan
| | - Tetsuya Kadonosono
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Shinae Kizaka-Kondoh
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan.
| |
Collapse
|
11
|
Yang Y, Wang C, Li Z, Lu Q, Li Y. Precise diagnosis and treatment of non-muscle invasive bladder cancer - A clinical perspective. Front Oncol 2023; 13:1042552. [PMID: 36798814 PMCID: PMC9927396 DOI: 10.3389/fonc.2023.1042552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
According to the guidelines, transurethral resection of bladder tumor (TURBT) followed by intravesical therapy remains the standard strategy for the management of non-muscle invasive bladder cancer (NMIBC). However, even if patients receive standard strategy, the risk of postoperative recurrence and progression is high. From the clinical perspective, the standard strategy needs to be optimized and improved. Compared to conventional TURBT, the technique of en bloc resection of bladder tumor (ERBT) removes the tumor tissue in one piece, thus following the principles of cancer surgery. Meanwhile, the integrity and spatial orientation of tumor tissue is protected during the operation, which is helpful for pathologists to make accurate histopathological analysis. Then, urologists can make a postoperative individualized treatment plan based on the patient's clinical characteristics and histopathological results. To date, there is no strong evidence that NMIBC patients treated with ERBT achieve better oncological prognosis, which indicates that ERBT alone does not yet improve patient outcomes. With the development of enhanced imaging technology and proteogenomics technology, en bloc resection combined with these technologies will make it possible to achieve precise diagnosis and treatment of bladder cancer. In this review, the authors analyze the current existing shortcomings of en bloc resection and points out its future direction, in order to promote continuous optimization of the management strategy of bladder cancer.
Collapse
Affiliation(s)
| | | | | | - Qiang Lu
- *Correspondence: Qiang Lu, ; Yuanwei Li,
| | - Yuanwei Li
- *Correspondence: Qiang Lu, ; Yuanwei Li,
| |
Collapse
|
12
|
Singhal S, Azari F, Caponetti GC, Kennedy GT. Novel intraoperative near-infrared imaging strategy to identify abnormalities in the anterior mediastinum. J Cardiothorac Surg 2022; 17:302. [PMID: 36494869 PMCID: PMC9734605 DOI: 10.1186/s13019-022-02054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Thoracic surgeons are frequently asked to biopsy suspicious tissues in the anterior mediastinum to discriminate between a reactive versus malignant pathology such as lymph nodes. The most common benign cause of a mediastinal lymph node is a reactive lymph node from a prior infection or inflammatory process such as post-COVID or granulomatous disease. The most common malignant cause is a lymphoproliferative disorder but also metastatic disease from neck, breast and other regional cancers. Biopsies in this location are challenging because they are far from the trachea and the sternum is a barrier to most diagnostic procedures. Thus, a surgical biopsy is frequently required and a common procedure for Thoracic surgeons. Technically, identifying these lesions can be challenging, particularly for small lesions or those in patients with high body mass index. In order to improve contrast between diseased tissue in the anterior mediastinum and surrounding adipose tissue, we have been studying near-infrared imaging during surgery using indocyanine green (ICG) to give contrast to the abnormal tissues and to avoid an unnecessary extended resection. We developed a modified technique to give ICG to a patient during a biopsy in the anterior mediastinum to specifically highlight abnormal tissues. As a proof-of-principle, we present a case of a young woman with a suspicious 2 cm mediastinal lymph node that required surgical biopsy.
Collapse
Affiliation(s)
- Sonia Singhal
- grid.25879.310000 0004 1936 8972Department of Surgery, University of Pennsylvania Perelman School of Medicine, Pennsylvania, PA USA
| | - Feredun Azari
- grid.25879.310000 0004 1936 8972Department of Surgery, University of Pennsylvania Perelman School of Medicine, Pennsylvania, PA USA
| | - Gabriel C. Caponetti
- grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Pennsylvania, PA USA
| | - Gregory T. Kennedy
- grid.25879.310000 0004 1936 8972Department of Surgery, University of Pennsylvania Perelman School of Medicine, Pennsylvania, PA USA ,grid.411115.10000 0004 0435 0884Department of Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104 USA
| |
Collapse
|
13
|
Kim H, Kim J, Sa JK, Ryu BK, Park KJ, Kim J, Ha H, Park Y, Shin MH, Kim J, Lee H, Kim D, Lee K, Jang B, Lee KM, Kang SH. Calcipotriol, a synthetic Vitamin D analog, promotes antitumor immunity via CD4+T-dependent CTL/NK cell activation. Biomed Pharmacother 2022; 154:113553. [PMID: 35994815 DOI: 10.1016/j.biopha.2022.113553] [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: 07/08/2022] [Revised: 08/09/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022] Open
Abstract
To overcome the hurdles of immunotherapy, we investigated whether calcipotriol, a synthetic vitamin D analog, could overcome the immune evasion of glioblastoma multiforme (GBM) by modulating immune responses and the immunosuppressive tumor microenvironment. Administration of calcipotriol considerably reduced tumor growth. Both in vivo and in vitro studies revealed that CD8+T and natural killer (NK) cell gene signatures were enriched and activated, producing high levels of IFN-γ and granzyme B. In contrast, regulatory T cells (Treg) were significantly reduced in the calcipotriol-treated group. The expression of CD127, the receptor for thymic stromal lymphopoietin (TSLP), is elevated in CD4+T cells and potentially supports T-cell priming. Depleting CD4+T cells, but not NK or CD8+T cells, completely abrogated the antitumor efficacy of calcipotriol. These data highlight that the calcipotriol/TSLP/CD4+T axis can activate CD8+T and NK cells with a concomitant reduction in the number of Tregs in GBM. Therefore, calcipotriol can be a novel therapeutic modality to overcome the immune resistance of GBM by converting immunologically "cold" tumors into "hot" tumors. DATA AVAILABILITY: Data are available upon reasonable request. The RNA-seq dataset comparing the transcriptomes of control and calcipotriol-treated GL261 tumors is available from the corresponding author upon request.
Collapse
Affiliation(s)
- Hyungsin Kim
- Department of Neurosurgery, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Jeongsoo Kim
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Jason K Sa
- Biomedical Sciences, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Byung-Kyu Ryu
- Department of Neurosurgery, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Kyung-Jae Park
- Department of Neurosurgery, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Jiyoung Kim
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Hyojeong Ha
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Yejin Park
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Min Hwa Shin
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Jungwon Kim
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Hyemin Lee
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Daham Kim
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Kyunghye Lee
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Byunghyun Jang
- Biomedical Sciences, Korea University College of Medicine, Seoul, the Republic of Korea
| | - Kyung-Mi Lee
- Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, the Republic of Korea.
| | - Shin-Hyuk Kang
- Department of Neurosurgery, Korea University College of Medicine, Seoul, the Republic of Korea.
| |
Collapse
|
14
|
Muto J, Mine Y, Nishiyama Y, Murayama K, Yamada S, Kojima D, Hayakawa M, Adachi K, Hasegawa M, Lee JYK, Hirose Y. Intraoperative Real-Time Near-Infrared Image-Guided Surgery to Identify Intracranial Meningiomas via Microscope. Front Neurosci 2022; 16:837349. [PMID: 35600609 PMCID: PMC9114498 DOI: 10.3389/fnins.2022.837349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/24/2022] Open
Abstract
Meningiomas are a common pathology in the central nervous system requiring complete surgical resection. However, in cases of recurrence and post-irradiation, accurate identification of tumor remnants and a dural tail under bright light remains challenging. We aimed to perform real-time intraoperative visualization of the meningioma and dural tail using a delayed-window indocyanine green (ICG) technique with microscopy. Fifteen patients with intracranial meningioma received 0.5 mg/kg ICG a few hours before observation during the surgery. We used near-infrared (NIR) fluorescence to identify the tumor location. NIR fluorescence could visualize meningiomas in 12 out of 15 cases. Near-infrared visualization during the surgery ranged from 1 to 4 h after the administration of ICG. The mean signal-to-background ratio (SBR) of the intracranial meningioma in delayed-window ICG (DWIG) was 3.3 ± 2.6. The ratio of gadolinium-enhanced T1 tumor signal to the brain (T1BR) (2.5 ± 0.9) was significantly correlated with the tumor SBR (p = 0.016). Ktrans, indicating blood–brain barrier permeability, was significantly correlated with tumor SBR (p < 0.0001) and T1BR (p = 0.013) on dynamic contrast-enhanced magnetic resonance imaging (MRI). DWIG demonstrated a sensitivity of 94%, specificity of 38%, positive predictive value (PPV) of 76%, and negative predictive value (NPV) of 75% for meningiomas. This is the first pilot study in which DWIG fluorescence-guided surgery was used to visualize meningioma and dural tail intraoperatively with microscopy. DWIG is comparable with second-window ICG in terms of mean SBR. Gadolinium-enhanced T1 tumor signal may predict NIR fluorescence of the intracranial meningioma. Blood–brain barrier permeability as shown by Ktrans on dynamic contrast-enhanced MRI can contribute to gadolinium enhancement on MRI and to ICG retention and tumor fluorescence by NIR.
Collapse
Affiliation(s)
- Jun Muto
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
- *Correspondence: Jun Muto,
| | - Yutaka Mine
- Department of Neurosurgery, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan
| | - Yuya Nishiyama
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | | | - Seiji Yamada
- Department of Pathology, Fujita Health University, Toyoake, Japan
| | - Daijiro Kojima
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | - Motoharu Hayakawa
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | - Kazuhide Adachi
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| | | | - John Y. K. Lee
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University, Toyoake, Japan
| |
Collapse
|
15
|
CD47-targeted optical molecular imaging and near-infrared photoimmunotherapy in the detection and treatment of bladder cancer. Mol Ther Oncolytics 2022; 24:319-330. [PMID: 35118190 PMCID: PMC8784304 DOI: 10.1016/j.omto.2021.12.020] [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: 10/01/2021] [Accepted: 12/28/2021] [Indexed: 12/03/2022] Open
Abstract
Transurethral resection of bladder tumor (TURBT) followed by intravesical therapy remains the most effective strategy for the management of non-muscle-invasive bladder cancer worldwide. TURBT has two purposes: to remove all visible tumors and to obtain tumor specimens for histopathological analysis. However, the detection of flat and small malignant lesions under white-light cystoscopy is extremely challenging, and residual lesions are still the main reason for the high recurrence rate of bladder cancer. We hypothesized that visual enhancement of malignant lesions using targeted optical molecular imaging could potentially highlight residual tumors in the bladder during surgery, and near-infrared photoimmunotherapy (NIR-PIT) could kill exfoliated cancer cells and residual tumors. A mouse model of complete or partial bladder tumor resection was established under the guidance of optical molecular imaging mediated by indocyanine green and anti-CD47-Alexa Fluor 790, respectively. Once the tumor recurred, mouse model received repeated CD47-targeted NIR-PIT. After complete resection, there was no tumor recurrence. Furthermore, the growth rate of recurrent tumor decreased significantly after repeated NIR-PIT. Therefore, CD47-targeted optical molecular imaging can potentially assist urologists to detect and remove all tumors, and repeated NIR-PIT shows the potential to reduce tumor recurrence rates and inhibit the growth of recurrent tumor.
Collapse
|
16
|
Muto J, Mine Y, Nagai S, Shizu N, Takeda H, Ikeda D, Saito A, Joko M, Hasegawa M, Kaneko S, Inoue T, Lee JYK, Hirose Y. Utility of intraoperative real-time near-infrared fluorescence surgery for spinal schwannoma. NEUROSURGICAL FOCUS: VIDEO 2022; 6:V12. [PMID: 36284589 PMCID: PMC9557329 DOI: 10.3171/2021.10.focvid21158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 10/25/2021] [Indexed: 11/29/2022]
Abstract
The authors report the first cases of fluorescence-guided spinal surgery of schwannomas using near-infrared fluorescence imaging with the delayed window indocyanine (ICG) green (DWIG) technique for accurate real-time intraoperative tumor visualization. Patients with intradural spinal schwannomas received 0.5 mg/kg ICG at the beginning of surgery. After 1 hour, using the DWIG technique, near-infrared spectroscopy (NIRS) detected the spinal schwannomas, showing the exact tumor location and boundaries. DWIG with NIRS microscopy confirmed the exact location of spinal schwannomas before and after opening of the dura mater, thereby facilitating successful tumor dissection from the surrounding tissues, tumor resection, and confirmation of tumor removal. The video can be found here: https://stream.cadmore.media/r10.3171/2021.10.FOCVID21158
Collapse
Affiliation(s)
- Jun Muto
- Department of Neurosurgery, Fujita Health University, Toyoake
| | - Yutaka Mine
- Department of Neurosurgery, Brain Nerve Center, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Japan; and
| | - Sota Nagai
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake
| | - Naoyuki Shizu
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake
| | - Hiroki Takeda
- Department of Spine and Spinal Cord Surgery, Fujita Health University, Toyoake
| | - Daiki Ikeda
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake
| | - Akifumi Saito
- Department of Spine and Spinal Cord Surgery, Fujita Health University, Toyoake
| | - Masahiro Joko
- Department of Neurosurgery, Fujita Health University, Toyoake
| | | | - Shinjiro Kaneko
- Department of Spine and Spinal Cord Surgery, Fujita Health University, Toyoake
| | - Tatsushi Inoue
- Department of Neurosurgery, Fujita Health University, Toyoake
| | - John Y. K. Lee
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yuichi Hirose
- Department of Neurosurgery, Fujita Health University, Toyoake
| |
Collapse
|
17
|
Li Y, You Q, Wang Z, Cao Y, Butch CJ, Guissi NEI, Cai H, Wang Y, Lu Q. A study on setting standards for near-infrared fluorescence-image guided surgery (NIRFGS) time lapse monitoring based on preoperative liver function assessment. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:96. [PMID: 35282106 PMCID: PMC8848407 DOI: 10.21037/atm-21-6975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/21/2022] [Indexed: 11/06/2022]
Abstract
Background This study aimed to explore the relationship between the fluorescence intensity of indocyanine green (ICG) in near-infrared fluorescence guided surgery (NIRFGS) and preoperative liver function indicators. Methods A total of 12 4T1 tumor-bearing mice were used for model establishment. Intraperitoneal injection (i.p.) of 20% carbon tetrachloride (CCl4) corn oil solution (50 µL) was given to mice in the liver injury model group, 24 hours after injection, the model was established, while the control group received 0% CCl4 corn oil solution (50 µL) (n=6 for each group). Additionally, doses of 8 mg/kg and 1 mg/kg of free ICG were injected intravenously (i.v.) (n=3 in each group). Fluorescence was imaged in vivo using an NIR fluorescence imaging system at different time points (1, 2, 4, 8, 12, 24, 48, and 72 h) after injection. Results The absolute fluorescence intensity of mice in the liver injury model group was stronger than that in the control group. Mice in the liver injury model group had the same clearance rate of ICG from the tumor as normal mice. However, the background clearance rate was slower than that of normal mice, which prolonged the optimal tumor to background ratio (TBR) time. Correlation analysis was also used to determine which preoperative liver function parameters were most correlated with hepatic ICG clearance. Conclusions Liver injury does not significantly affect the maximum TBR, but prolongs the optimal TBR time, and at the same time, a wider and more stable surgical window will appear. This study showed that a prolonged surgical start time is feasible according to preoperative liver function testing using NIR fluorescence imaging technology.
Collapse
Affiliation(s)
- Yunlong Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Qi You
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Ziyang Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Ying Cao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Christopher J Butch
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Nida El Islem Guissi
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Huiming Cai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.,Department of Research and Development Center, Nanjing Nuoyuan Medical Devices Co. Ltd., Nanjing, China
| | - Yiqing Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Qian Lu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.,Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| |
Collapse
|
18
|
Brookes MJ, Chan CD, Nicoli F, Crowley TP, Ghosh KM, Beckingsale T, Saleh D, Dildey P, Gupta S, Ragbir M, Rankin KS. Intraoperative Near-Infrared Fluorescence Guided Surgery Using Indocyanine Green (ICG) for the Resection of Sarcomas May Reduce the Positive Margin Rate: An Extended Case Series. Cancers (Basel) 2021; 13:cancers13246284. [PMID: 34944902 PMCID: PMC8699240 DOI: 10.3390/cancers13246284] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/03/2021] [Accepted: 12/11/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Sarcomas are rare, aggressive cancers which can occur in any region of the body. Surgery is usually the cornerstone of curative treatment, with negative surgical margins associated with decreased local recurrence and improved overall survival. Indocyanine green (ICG) is a fluorescent dye which accumulates in sarcoma tissue and can be imaged intraoperatively using handheld near-infrared (NIR) cameras, theoretically helping guide the surgeon's resection margins. METHODS Patients operated on between 20 February 2019 and 20 October 2021 for intermediate to high grade sarcomas at our centres received either conventional surgery, or were administered ICG pre-operatively followed by intra-operative NIR fluorescence guidance during the procedure. Differences between the unexpected positive margin rates were compared. RESULTS 115 suitable patients were identified, of which 39 received ICG + NIR fluorescence guided surgery, and 76 received conventional surgery. Of the patients given ICG, 37/39 tumours fluoresced, and surgeons felt the procedure was guided by the intra-operative images in 11 cases. Patients receiving ICG had a lower unexpected positive margin rate (5.1% vs. 25.0%, p = 0.01). CONCLUSIONS The use of NIR fluorescence cameras in combination with ICG may reduce the unexpected positive margin rate for high grade sarcomas. A prospective, multi-centre randomised control trial is now needed to validate these results.
Collapse
Affiliation(s)
- Marcus J. Brookes
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Correspondence: (M.J.B.); (K.S.R.)
| | - Corey D. Chan
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Fabio Nicoli
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
| | - Timothy P. Crowley
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
| | - Kanishka M. Ghosh
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
| | - Thomas Beckingsale
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
| | - Daniel Saleh
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
| | - Petra Dildey
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
| | - Sanjay Gupta
- Department of Musculoskeletal Oncology Surgery, Glasgow Royal Infirmary, 84 Castle St., Glasgow G4 0SF, UK;
| | - Maniram Ragbir
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
| | - Kenneth S. Rankin
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne NE1 4LP, UK; (C.D.C.); (F.N.); (T.P.C.); (K.M.G.); (T.B.); (D.S.); (P.D.); (M.R.)
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
- Correspondence: (M.J.B.); (K.S.R.)
| |
Collapse
|
19
|
Enhanced Detection of Desmoplasia by Targeted Delivery of Iron Oxide Nanoparticles to the Tumour-Specific Extracellular Matrix. Pharmaceutics 2021; 13:pharmaceutics13101663. [PMID: 34683956 PMCID: PMC8539756 DOI: 10.3390/pharmaceutics13101663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 01/14/2023] Open
Abstract
Diagnostic imaging of aggressive cancer with a high stroma content may benefit from the use of imaging contrast agents targeted with peptides that have high binding affinity to the extracellular matrix (ECM). In this study, we report the use of superparamagnetic iron-oxide nanoparticles (IO-NP) conjugated to a nonapeptide, CSGRRSSKC (CSG), which specifically binds to the laminin-nidogen-1 complex in tumours. We show that CSG-IO-NP accumulate in tumours, predominantly in the tumour ECM, following intravenous injection into a murine model of pancreatic neuroendocrine tumour (PNET). In contrast, a control untargeted IO-NP consistently show poor tumour uptake, and IO-NP conjugated to a pentapeptide, CREKA that bind fibrin clots in blood vessels show restricted uptake in the angiogenic vessels of the tumours. CSG-IO-NP show three-fold higher intratumoral accumulation compared to CREKA-IO-NP. Magnetic resonance imaging (MRI) T2-weighted scans and T2 relaxation times indicate significant uptake of CSG-IO-NP irrespective of tumour size, whereas the uptake of CREKA-IO-NP is only consistent in small tumours of less than 3 mm in diameter. Larger tumours with significantly reduced tumour blood vessels show a lack of CREKA-IO-NP uptake. Our data suggest CSG-IO-NP are particularly useful for detecting stroma in early and advanced solid tumours.
Collapse
|
20
|
Gangadharan S, Sarkaria IN, Rice D, Murthy S, Braun J, Kucharczuk J, Predina J, Singhal S. Multiinstitutional Phase 2 Clinical Trial of Intraoperative Molecular Imaging of Lung Cancer. Ann Thorac Surg 2021; 112:1150-1159. [PMID: 33221195 PMCID: PMC10985531 DOI: 10.1016/j.athoracsur.2020.09.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Intraoperative molecular imaging (IMI) may improve surgical outcomes during pulmonary resection for lung cancer. A multiinstitutional phase 2 IMI clinical trial was conducted using a near-infrared, folate receptor-targeted contrast agent for lung adenocarcinomas, OTL38. The primary goal was to determine whether OTL38 improved surgeons' ability to identify difficult to find nodules, occult cancers, and positive margins. METHODS Patients with lung nodules received OTL38 (0.025 mg/kg) preoperatively. Patients had IMI sequentially during lung inspection, tumor resection, and margin check. Efficacy was evaluated by occurrence of clinically significant events, occurrences that caused the surgeon to modify the operation or upstage the patient's cancer. Safety was assessed for a single intravenous dose of OTL38. RESULTS Of 110 patients recruited, 92 were eligible for analysis. During lung inspection, IMI found 24 additional nodules, 9 (10%) of which were cancers that had not been known preoperatively. During tumor resection, IMI located 11 (12%) lesions that the surgeon could not find. During the margin check, IMI revealed 8 positive margins (9%) that the surgeon thought were negative. Benefits of IMI were pronounced in patients undergoing sublobar pulmonary resections and in patients with ground-glass opacities. There were no serious adverse events. All surgeons felt comfortable with the procedures by 10 cases. CONCLUSIONS In this phase 2 clinical trial, IMI improved outcomes for 26% of patients. A randomized, multiinstitutional phase 3 clinical trial is underway.
Collapse
Affiliation(s)
- Sidhu Gangadharan
- Division of Thoracic Surgery, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Inderpal N Sarkaria
- Division of Thoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David Rice
- Division of Thoracic Surgery, MD Anderson Cancer Center, Houston, Texas
| | - Sudish Murthy
- Division of Thoracic Surgery, Cleveland Clinic, Cleveland, Ohio
| | - Jerry Braun
- Division of Thoracic Surgery, University of Leiden, Leiden, the Netherlands
| | - John Kucharczuk
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Jarrod Predina
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
21
|
Cao Y, Wang P, Wang Z, Zhang W, Lu Q, Butch CJ, Guissi NEI, You Q, Cai H, Ding Y, Wang Y. A pilot study of near-infrared fluorescence guided surgery for primary tumor localization and lymph node mapping in colorectal cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1342. [PMID: 34532479 PMCID: PMC8422097 DOI: 10.21037/atm-21-4021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/20/2021] [Indexed: 01/05/2023]
Abstract
Background This pilot study aimed to evaluate the feasibility of near-infrared fluorescence imaging for primary tumor localization, lymph node mapping, and metastatic lymph node detection in colorectal cancer (CRC) using indocyanine green (ICG). Methods A total of 11 patients with CRC were prospectively enrolled. ICG (25 mg dissolved in 30 mL sterile water) was intravenously injected preoperatively, and the fluorescence intensity of the primary tumor, lymph nodes, and normal tissues, as well as the signal-to-background ratio (SBR) and contrast-to-noise ratio (CNR) were measured at 0.5, 1, 2, 4, and 24 h after ICG injection. Results The primary tumor could be located intraoperatively, and the tumor boundary was clear at 2–4 h. There was good contrast in the fluorescence intensity between tumor and normal tissues (SBR =2.11±0.36, CNR =8.74±0.35). The lymph node detection rate was 95% (38/40), and the SBR threshold of lymph nodes was 1.13. Conclusions This pilot study showed that primary tumor localization and lymph node mapping in CRC is feasible using near-infrared fluorescence imaging technology, though metastatic lymph nodes cannot be discriminated from benign ones. In addition, cancer nodules missed by both white light mode and palpation by the surgeon were unexpectedly found, resulting in a change in the surgical prognosis in 9.1% (1/11) of patients.
Collapse
Affiliation(s)
- Ying Cao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Peng Wang
- Department of General Surgery, Pukou Branch of Jiangsu People's Hospital, Nanjing, China.,Department of general surgery, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Ziyang Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Wei Zhang
- Jiangsu Testing and Inspection Institute for Medical Devices, Nanjing, China
| | - Qian Lu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Christopher J Butch
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Nida El Islem Guissi
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Qi You
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| | - Huiming Cai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.,Nanjing Nuoyuan Medical Devices Co., Ltd, Nanjing, China
| | - Yongbin Ding
- Department of General Surgery, Pukou Branch of Jiangsu People's Hospital, Nanjing, China
| | - Yiqing Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
| |
Collapse
|
22
|
Cho SS, Salinas R, De Ravin E, Teng CW, Li C, Abdullah KG, Buch L, Hussain J, Ahmed F, Dorsey J, Mohan S, Brem S, Singhal S, Lee JYK. Near-Infrared Imaging with Second-Window Indocyanine Green in Newly Diagnosed High-Grade Gliomas Predicts Gadolinium Enhancement on Postoperative Magnetic Resonance Imaging. Mol Imaging Biol 2021; 22:1427-1437. [PMID: 31712948 DOI: 10.1007/s11307-019-01455-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Intraoperative molecular imaging with tumor-targeting fluorophores offers real-time detection of neoplastic tissue. The second window indocyanine green (SWIG) technique relies on passive accumulation of indocyanine green (ICG), a near-infrared fluorophore, in neoplastic tissues. In this study, we explore the ability of SWIG to detect neoplastic tissue and to predict postoperative magnetic resonance imaging (MRI) findings intraoperatively. PROCEDURES Retrospective data were collected from 36 patients with primary high-grade gliomas (HGG) enrolled as part of a larger trial between October 2014 and October 2018. Patients received systemic ICG infusions at 2.5-5 mg/kg 24 h preoperatively. Near-infrared fluorescence was recorded throughout the case and from biopsy specimens. The presence/location of residual SWIG signal after resection was compared to the presence/location of residual gadolinium enhancement on postoperative MRI. The extent of resection was not changed based on near-infrared imaging. RESULTS All 36 lesions demonstrated strong near-infrared fluorescence (signal-to-background = 6.8 ± 2.2) and 100 % of tumors reaching the cortex were visualized before durotomy. In 78 biopsy specimens, near-infrared imaging demonstrated higher sensitivity and accuracy than white light for diagnosing neoplastic tissue intraoperatively. Furthermore, near-infrared imaging predicted gadolinium enhancement on postoperative MRI with 91 % accuracy, with visualization of residual enhancement as small as 0.3 cm3. Patients with no residual near-infrared signal after resection were significantly more likely to have complete resection on postoperative MRI (p value < 0.0001). CONCLUSIONS Intraoperative imaging with SWIG demonstrates highly sensitive detection of HGG tissue in real time. Furthermore, post-resection near-infrared imaging correlates with postoperative MRI. Overall, our findings suggest that SWIG can provide surgeons with MRI-like results in real time, potentially increasing resection rates.
Collapse
Affiliation(s)
- Steve S Cho
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Salinas
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Emma De Ravin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Clare W Teng
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Carrie Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Love Buch
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jasmin Hussain
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Fahad Ahmed
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jay Dorsey
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Brem
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Bryski MG, Frenzel-Sulyok LG, Delikatny EJ, Deshpande C, Litzky LA, Singhal S. Molecular imaging can identify the location to perform a frozen biopsy during intraoperative frozen section consultation. PLoS One 2021; 16:e0252731. [PMID: 34086790 PMCID: PMC8177495 DOI: 10.1371/journal.pone.0252731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/20/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Intraoperative frozen section (FS) consultation is an important tool in surgical oncology that suffers from sampling error because the pathologist does not always know where to perform a biopsy of the surgical specimen. Intraoperative molecular imaging is a technology used in the OR to visualize lesions during surgery. We hypothesized that molecular imaging can address this pathology challenge in FS by visualizing the cancer cells in the specimen in the pathology suite. Here, we report the development and validation of a molecular-imaging capable cryostat called Smart-Cut. METHODS A molecular imaging capable cryostat prototype was developed and tested using a murine model. Tumors grown in mice were targeted with a NIR contrast agent, indocyanine green (ICG), via tail vein injection. Tumors and adjacent normal tissue samples were frozen sectioned with Smart-Cut. Fluorescent sections and non-fluorescent sections were prepared for H&E and fluorescent microscopy. Fluorescent signal was quantified by tumor-to-background ratio (TBR). NIR fluorescence was tested in one patient enrolled in a clinical trial. RESULTS The Smart-Cut prototype has a small footprint and fits well in the pathology suite. Fluorescence imaging with Smart-Cut identified cancerous tissue in the specimen in all 12 mice. No false positives or false negatives were seen, as confirmed by H&E. The mean TBR in Smart-Cut positive tissue sections was 6.8 (SD±3.8). In a clinical application in the pathology suite, NIR imaging identified two lesions in a pulmonary resection specimen, where traditional grossing only identified one. CONCLUSION Molecular imaging can be integrated into the pathology suite via the Smart-Cut device, and can detect cancer in frozen tissue sections using molecular imaging in a murine model.
Collapse
Affiliation(s)
- Mitchell G. Bryski
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lydia G. Frenzel-Sulyok
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - E. James Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Charuhas Deshpande
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Leslie A. Litzky
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sunil Singhal
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
24
|
Fluorophores Use in Pituitary Surgery: A Pharmacokinetics and Pharmacodynamics Appraisal. Brain Sci 2021; 11:brainsci11050565. [PMID: 33925235 PMCID: PMC8146254 DOI: 10.3390/brainsci11050565] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/15/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
(1) Background: Despite many surgical and technological advances, pituitary adenoma surgery is still burdened by non-negligible rates of incomplete tumor resection, mainly due to difficulties in differentiating pathology from normal pituitary tissue. Some fluorescent agents have been recently investigated as intraoperative contrast agents in pituitary surgery. The aim of this study is to evaluate the actual knowledge about the usefulness of such fluorophores with a particular focus on both the pharmacokinetics and pharmacodynamics issues of the pituitary gland. (2) Methods: We reviewed the current literature about fluorophores use in pituitary surgery and reported the first fully endoscopic experience with fluorescein. (3) Results: The studies investigating 5-ALA use reported contrasting results. ICG showed encouraging results, although with some specificity issues in identifying pathological tissue. Low-dose fluorescein showed promising results in differentiating pathology from normal pituitary tissue. Apart from the dose and timing of administration, both the fluorophores' volume of distribution and the histological variability of the interstitial space and vascular density played a crucial role in optimizing intraoperative contrast enhancement. (4) Conclusions: Both pharmacokinetics and pharmacodynamics issues determine the potential usefulness of fluorophores in pituitary surgery. ICG and fluorescein showed the most promising results, although further studies are needed.
Collapse
|
25
|
Bourgeois P, Veys I, Noterman D, De Neubourg F, Chintinne M, Vankerckhove S, Nogaret JM. Near-Infrared Fluorescence Imaging of Breast Cancer and Axillary Lymph Nodes After Intravenous Injection of Free Indocyanine Green. Front Oncol 2021; 11:602906. [PMID: 33767980 PMCID: PMC7985064 DOI: 10.3389/fonc.2021.602906] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/28/2021] [Indexed: 11/13/2022] Open
Abstract
Background Near-infrared fluorescence imaging (NIRFI) of breast cancer (BC) after the intravenous (IV) injection of free indocyanine green (fICG) has been reported to be feasible. However, some questions remained unclarified. Objective To evaluate the distribution of fICG in BC and the axillary lymph nodes (LNs) of women undergoing surgery with complete axillary LN dissection (CALND) and/or selective lymphadenectomy (SLN) of sentinel LNs (NCT no. 01993576 and NCT no. 02027818). Methods An intravenous injection of fICG (0.25 mg/kg) was administered to one series of 20 women undergoing treatment with mastectomy, the day before surgery in 5 (group 1) and immediately before surgery in 15 (group 2: tumor localization, 25; and pN+ CALND, 4) as well as to another series of 20 women undergoing treatment with tumorectomy (group 3). A dedicated NIR camera was used for ex vivo fluorescence imaging of the 45 BC lesions and the LNs. Results In group 1, two of the four BC lesions and one large pN+ LN exhibited fluorescence. In contrast, 24 of the 25 tumors in group 2 and all of the tumors in group 3 were fluorescent. The sentinel LNs were all fluorescent, as well as some of the LNs in all CALND specimens. Metastatic cells were found in the fluorescent LNs of the pN+ cases. Fluorescent BC lesions could be identified ex vivo on the surface of the lumpectomy specimen in 14 of 19 cases. Conclusions When fICG is injected intravenously just before surgery, BC can be detected using NIRFI with high sensitivity, with metastatic axillary LNs also showing fluorescence. Such a technical approach seems promising in the management of BC and merits further investigation.
Collapse
Affiliation(s)
- Pierre Bourgeois
- Nuclear Medicine Service, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Veys
- Surgery Service, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Danielle Noterman
- Surgery Service, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Filip De Neubourg
- Surgery Service, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Marie Chintinne
- Department of Anatomo-Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Sophie Vankerckhove
- Nuclear Medicine Service, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Marie Nogaret
- Surgery Service, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
26
|
Wang Z, Chen M, Liu JJ, Chen RH, Yu Q, Wang GM, Nie LM, Huang WH, Zhang GJ. Human Serum Albumin Decorated Indocyanine Green Improves Fluorescence-Guided Resection of Residual Lesions of Breast Cancer in Mice. Front Oncol 2021; 11:614050. [PMID: 33763353 PMCID: PMC7983674 DOI: 10.3389/fonc.2021.614050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/28/2021] [Indexed: 11/13/2022] Open
Abstract
Objective Achieving negative resection margin is critical but challenging in breast-conserving surgery. Fluorescence-guided surgery allows the surgeon to visualize the tumor bed in real-time and to facilitate complete resection. We envisioned that intraoperative real-time fluorescence imaging with a human serum albumin decorated indocyanine green probe could enable complete surgical removal of breast cancer in a mouse model. Methods We prepared the probe by conjugating indocyanine green (ICG) with human serum albumin (HSA). In vitro uptake of the HSA-ICG probe was compared between human breast cancer cell line MDA-MB-231 and normal breast epithelial cell line MCF 10A. In vivo probe selectivity for tumors was examined in nude mice bearing MDA-MB-231-luc xenografts and the FVB/N-Tg (MMTV-PyMT) 634Mul/J mice model with spontaneous breast cancer. A positive-margin resection mice model bearing MDA-MB-231-luc xenograft was established and the performance of the probe in assisting surgical resection of residual lesions was examined. Results A significantly stronger fluorescence intensity was detected in MDA-MB-231 cells than MCF 10A cells incubated with HSA-ICG. In vivo fluorescence imaging showed that HSA-ICG had an obvious accumulation at tumor site at 24 h with tumor-to-normal tissue ratio of 8.19 ± 1.30. The same was true in the transgenic mice model. The fluorescence intensity of cancer tissues was higher than that of non-cancer tissues (58.53 ± 18.15 vs 32.88 ± 11.34). During the surgical scenarios, the residual tumors on the surgical bed were invisible with the naked eye, but were detected and resected with negative margin under HSA-ICG guidance in all the mice (8/8). Recurrence rate among mice that underwent resection with HSA-ICG (0/8) was significantly lower than the rates among mice with ICG (4/8), as well as the control group under white light (7/7). Conclusions This study suggests that real-time in vivo visualization of breast cancer with an HSA-ICG fluorescent probe facilitates complete surgical resection of breast cancer in a mouse xenograft model.
Collapse
Affiliation(s)
- Zun Wang
- ChangJiang Scholar's Laboratory, Medical College, Shantou University, Shantou, China
| | - Min Chen
- Clinical Central Research Core, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.,Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, Xiang'an Hospital of Xiamen University, Xiamen, China.,Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Jing-Jing Liu
- Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, Xiang'an Hospital of Xiamen University, Xiamen, China.,Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.,Cancer Center & Department of Breast and Thyroid Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Rong-He Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Qian Yu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Gui-Mei Wang
- Department of Pathology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Li-Ming Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Wen-He Huang
- Cancer Center & Department of Breast and Thyroid Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Guo-Jun Zhang
- ChangJiang Scholar's Laboratory, Medical College, Shantou University, Shantou, China.,Key Laboratory for Endocrine-Related Cancer Precision Medicine of Xiamen, Xiang'an Hospital of Xiamen University, Xiamen, China.,Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.,Cancer Center & Department of Breast and Thyroid Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
27
|
Teng CW, Huang V, Arguelles GR, Zhou C, Cho SS, Harmsen S, Lee JYK. Applications of indocyanine green in brain tumor surgery: review of clinical evidence and emerging technologies. Neurosurg Focus 2021; 50:E4. [PMID: 33386005 DOI: 10.3171/2020.10.focus20782] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/23/2020] [Indexed: 11/06/2022]
Abstract
Indocyanine green (ICG) is a water-soluble dye that was approved by the FDA for biomedical purposes in 1956. Initially used to measure cardiocirculatory and hepatic functions, ICG's fluorescent properties in the near-infrared (NIR) spectrum soon led to its application in ophthalmic angiography. In the early 2000s, ICG was formally introduced in neurosurgery as an angiographic tool. In 2016, the authors' group pioneered a novel technique with ICG named second-window ICG (SWIG), which involves infusion of a high dose of ICG (5.0 mg/kg) in patients 24 hours prior to surgery. To date, applications of SWIG have been reported in patients with high-grade gliomas, meningiomas, brain metastases, pituitary adenomas, craniopharyngiomas, chordomas, and pinealomas.The applications of ICG have clearly expanded rapidly across different specialties since its initial development. As an NIR fluorophore, ICG has advantages over other FDA-approved fluorophores, all of which are currently in the visible-light spectrum, because of NIR fluorescence's increased tissue penetration and decreased autofluorescence. Recently, interest in the latest applications of ICG in brain tumor surgery has grown beyond its role as an NIR fluorophore, extending into shortwave infrared imaging and integration into nanotechnology. This review aims to summarize reported clinical studies on ICG fluorescence-guided surgery of intracranial tumors, as well as to provide an overview of the literature on emerging technologies related to the utility of ICG in neuro-oncological surgeries, including the following aspects: 1) ICG fluorescence in the NIR-II window; 2) ICG for photoacoustic imaging; and 3) ICG nanoparticles for combined diagnostic imaging and therapy (theranostic) applications.
Collapse
Affiliation(s)
- Clare W Teng
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and.,2Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vincent Huang
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and.,2Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gabriel R Arguelles
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and.,2Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cecilia Zhou
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and.,2Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steve S Cho
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and
| | - Stefan Harmsen
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and
| | - John Y K Lee
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and
| |
Collapse
|
28
|
Muto J, Mine Y, Nakagawa Y, Joko M, Kagami H, Inaba M, Hasegawa M, Lee JYK, Hirose Y. Intraoperative real-time near-infrared optical imaging for the identification of metastatic brain tumors via microscope and exoscope. Neurosurg Focus 2021; 50:E11. [PMID: 33386024 DOI: 10.3171/2020.10.focus20767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/22/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE As chemotherapy and radiotherapy have developed, the role of a neurosurgeon in the treatment of metastatic brain tumors is gradually changing. Real-time intraoperative visualization of brain tumors by near-infrared spectroscopy (NIRS) is feasible. The authors aimed to perform real-time intraoperative visualization of the metastatic tumor in brain surgery using second-window indocyanine green (SWIG) with microscope and exoscope systems. METHODS Ten patients with intraparenchymal brain metastatic tumors were administered 5 mg/kg indocyanine green (ICG) 1 day before the surgery. In some patients, a microscope was used to help identify the metastases, whereas in the others, an exoscope was used. RESULTS NIRS with the exoscope and microscope revealed the tumor location from the brain surface and the tumor itself in all 10 patients. The NIR signal could be detected though the normal brain parenchyma up to 20 mm. While the mean signal-to-background ratio (SBR) from the brain surface was 1.82 ± 1.30, it was 3.35 ± 1.76 from the tumor. The SBR of the tumor (p = 0.030) and the ratio of Gd-enhanced T1 tumor signal to normal brain (T1BR) (p = 0.0040) were significantly correlated with the tumor diameter. The SBR of the tumor was also correlated with the T1BR (p = 0.0020). The tumor was completely removed in 9 of the 10 patients, as confirmed by postoperative Gd-enhanced MRI. This was concomitant with the absence of NIR fluorescence at the end of surgery. CONCLUSIONS SWIG reveals the metastatic tumor location from the brain surface with both the microscope and exoscope systems. The Gd-enhanced T1 tumor signal may predict the NIR signal of the metastatic tumor, thus facilitating tumor resection.
Collapse
Affiliation(s)
- Jun Muto
- 1Department of Neurosurgery, Fujita Health University, Aichi
| | - Yutaka Mine
- 2Department of Neurosurgery, Saiseikai Yokohama Tobu Hospital, Kanagawa, Japan; and
| | - Yu Nakagawa
- 2Department of Neurosurgery, Saiseikai Yokohama Tobu Hospital, Kanagawa, Japan; and
| | - Masahiro Joko
- 1Department of Neurosurgery, Fujita Health University, Aichi
| | - Hiroshi Kagami
- 2Department of Neurosurgery, Saiseikai Yokohama Tobu Hospital, Kanagawa, Japan; and
| | - Makoto Inaba
- 2Department of Neurosurgery, Saiseikai Yokohama Tobu Hospital, Kanagawa, Japan; and
| | | | - John Y K Lee
- 3Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yuichi Hirose
- 1Department of Neurosurgery, Fujita Health University, Aichi
| |
Collapse
|
29
|
Teng CW, Amirshaghaghi A, Cho SS, Cai SS, De Ravin E, Singh Y, Miller J, Sheikh S, Delikatny E, Cheng Z, Busch TM, Dorsey JF, Singhal S, Tsourkas A, Lee JYK. Combined fluorescence-guided surgery and photodynamic therapy for glioblastoma multiforme using cyanine and chlorin nanocluster. J Neurooncol 2020; 149:243-252. [PMID: 32914293 PMCID: PMC7720701 DOI: 10.1007/s11060-020-03618-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) is the most common primary intracranial malignancy; survival can be improved by maximizing the extent-of-resection. METHODS A near-infrared fluorophore (Indocyanine-Green, ICG) was combined with a photosensitizer (Chlorin-e6, Ce6) on the surface of superparamagnetic-iron-oxide-nanoparticles (SPIONs), all FDA-approved for clinical use, yielding a nanocluster (ICS) using a microemulsion. The physical-chemical properties of the ICS were systematically evaluated. Efficacy of photodynamic therapy (PDT) was evaluated in vitro with GL261 cells and in vivo in a subtotal resection trial using a syngeneic flank tumor model. NIR imaging properties of ICS were evaluated in both a flank and an intracranial GBM model. RESULTS ICS demonstrated high ICG and Ce6 encapsulation efficiency, high payload capacity, and chemical stability in physiologic conditions. In vitro cell studies demonstrated significant PDT-induced cytotoxicity using ICS. Preclinical animal studies demonstrated that the nanoclusters can be detected through NIR imaging in both flank and intracranial GBM tumors (ex: 745 nm, em: 800 nm; mean signal-to-background 8.5 ± 0.6). In the flank residual tumor PDT trial, subjects treated with PDT demonstrated significantly enhanced local control of recurrent neoplasm starting on postoperative day 8 (23.1 mm3 vs 150.5 mm3, p = 0.045), and the treatment effect amplified to final mean volumes of 220.4 mm3 vs 806.1 mm3 on day 23 (p = 0.0055). CONCLUSION A multimodal theragnostic agent comprised solely of FDA-approved components was developed to couple optical imaging and PDT. The findings demonstrated evidence for the potential theragnostic benefit of ICS in surgical oncology that is conducive to clinical integration.
Collapse
Affiliation(s)
- Clare W Teng
- Department of Neurosurgery, Hospital of the University of Pennsylvania, 801 Spruce Street, 8th Floor, Philadelphia, PA, 19107, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmad Amirshaghaghi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Steve S Cho
- Department of Neurosurgery, Hospital of the University of Pennsylvania, 801 Spruce Street, 8th Floor, Philadelphia, PA, 19107, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuting S Cai
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Emma De Ravin
- Department of Neurosurgery, Hospital of the University of Pennsylvania, 801 Spruce Street, 8th Floor, Philadelphia, PA, 19107, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yash Singh
- Department of Neurosurgery, Hospital of the University of Pennsylvania, 801 Spruce Street, 8th Floor, Philadelphia, PA, 19107, USA
| | - Joann Miller
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saad Sheikh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Delikatny
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay F Dorsey
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, 801 Spruce Street, 8th Floor, Philadelphia, PA, 19107, USA.
| |
Collapse
|
30
|
Wojtynek NE, Olson MT, Bielecki TA, An W, Bhat AM, Band H, Lauer SR, Silva-Lopez E, Mohs AM. Nanoparticle Formulation of Indocyanine Green Improves Image-Guided Surgery in a Murine Model of Breast Cancer. Mol Imaging Biol 2020; 22:891-903. [PMID: 31820350 PMCID: PMC7280079 DOI: 10.1007/s11307-019-01462-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Negative surgical margins (NSMs) have favorable prognostic implications in breast tumor resection surgery. Fluorescence image-guided surgery (FIGS) has the ability to delineate surgical margins in real time, potentially improving the completeness of tumor resection. We have recently developed indocyanine green (ICG)-loaded self-assembled hyaluronic acid (HA) nanoparticles (NanoICG) for solid tumor imaging, which were shown to enhance intraoperative contrast. PROCEDURES This study sought to assess the efficacy of NanoICG on completeness of breast tumor resection and post-surgical survival. BALB/c mice bearing iRFP+/luciferase+ 4T1 syngeneic breast tumors were administered NanoICG or ICG, underwent FIGS, and were compared to bright light surgery (BLS) and sham controls. RESULTS NanoICG increased the number of complete resections and improved tumor-free survival. This was a product of improved intraoperative contrast enhancement and the identification of a greater number of small, occult lesions than ICG and BLS. Additionally, NanoICG identified chest wall invasion and predicted recurrence in a model of late-stage breast cancer. CONCLUSIONS NanoICG is an efficacious intraoperative contrast agent and could potentially improve surgical outcomes in breast cancer.
Collapse
Affiliation(s)
- Nicholas E Wojtynek
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Madeline T Olson
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Timothy A Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wei An
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aaqib M Bhat
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Scott R Lauer
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Edibaldo Silva-Lopez
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aaron M Mohs
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
31
|
Newton A, Predina J, Mison M, Runge J, Bradley C, Stefanovski D, Singhal S, Holt D. Intraoperative near-infrared imaging can identify canine mammary tumors, a spontaneously occurring, large animal model of human breast cancer. PLoS One 2020; 15:e0234791. [PMID: 32555698 PMCID: PMC7299356 DOI: 10.1371/journal.pone.0234791] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/02/2020] [Indexed: 01/05/2023] Open
Abstract
Introduction Current methods of intraoperative margin assessment in breast conserving surgery are impractical, unreliable, or time consuming. We hypothesized that intraoperative near-infrared (NIR) imaging with an FDA-approved NIR optical contrast agent could identify canine mammary tumors, a spontaneous large animal model of human breast cancer, during surgery. Methods Dogs with mammary tumors underwent a standard of care lumpectomy or mastectomy with wide surgical margins 20 hours after indocyanine green administration (3 mg/kg IV). During surgery, NIR imaging was performed on tumors and wound margins in situ and tumors and lymph nodes ex vivo. Following resection, the wound bed was examined for residual fluorescence. Fluorescence intensity was determined by signal-to-background ratio (SBR). All tumors, areas of residual fluorescence, and lymph nodes underwent histopathologic analysis. Results There were 41 mammary tumors in 16 female dogs. Twenty tumors were malignant and 21 were benign. Twenty-eight tumors were fluorescent (mean SBR 1.5±0.2). Sensitivity of fluorescence for all malignant tumors was 80% (16/20) and 93.3% (14/15) for malignant tumors > 2 cm. Specificity for malignancy was low (< 2cm = 55%; > 2cm = 30%). Tumors > 2 cm were more likely to be fluorescent (OR 6.05, 95% CI 1.50–24.44, P = 0.011) but not more likely to be malignant (OR 3.09, 95% CI 0.86–11.14, P = 0.085) than tumors ≤ 2 cm. Four out of seven inguinal lymph nodes excised in the mastectomy specimen fluoresced. All four drained malignant tumors; however only 2/4 contained metastatic disease. Conclusion Systemic ICG accumulates reliably in malignant canine mammary tumors > 2 cm. Although no tumor margins fluoresced, a wider margin of normal tissue is removed in canine mastectomy, making direct comparisons with breast conserving surgery difficult. Targeted NIR imaging agents are likely required to improve detection of smaller tumors and improve the specificity of NIR imaging for residual disease and metastatic lymph node detection.
Collapse
Affiliation(s)
- Andrew Newton
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jarrod Predina
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michael Mison
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jeffrey Runge
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Charles Bradley
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Darko Stefanovski
- Department of Clinical Studies New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - David Holt
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
32
|
Schwarz J, Schmidt H. Technology for Intraoperative Margin Assessment in Breast Cancer. Ann Surg Oncol 2020; 27:2278-2287. [PMID: 32350717 DOI: 10.1245/s10434-020-08483-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND As breast-conserving surgery (BCS) has become standard for treatment of breast cancer, the need for new technology to improve intraoperative margin assessment (IMA) has become clear. Close or positive margins during BCS lead to additional surgeries, treatment delay, additional stress for patients, and healthcare cost. Academia and industry have developed a diverse field of new technologies to allow surgeons to assess margins in the operating room. These technologies aim to reduce current rates of positive margins on final pathology. METHODS We selected recently developed IMA technologies, some of which have undergone large clinical trials and others that are still in early stage development. Technologies were categorized based on underlying methodology to differentiate malignant and normal tissue: spectroscopy, electrical properties, optical imaging and molecular imaging. Additionally, this review details clinical investigations, relevant statistical analysis as well as strengths and weaknesses of the various technologies. CONCLUSION Numerous technical innovations are being implemented to diminish rates of positive margins at breast tumor resection. Close collaboration among cross-disciplinary teams to further develop many of these technologies as well as completion of larger scale clinical studies are required to define an optimal approach. Development with an eye toward prioritizing sensitivity/specificity as well as healthcare cost containment has the potential to make a significant impact on this ongoing clinical need in breast cancer surgery.
Collapse
Affiliation(s)
- Julia Schwarz
- Dubin Breast Center, Tisch Cancer Institute, New York, USA.,Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Breast Surgical Oncology, The Mount Sinai Hospital, New York, NY, USA
| | - Hank Schmidt
- Dubin Breast Center, Tisch Cancer Institute, New York, USA. .,Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Breast Surgical Oncology, The Mount Sinai Hospital, New York, NY, USA.
| |
Collapse
|
33
|
Favril S, Abma E, Stock E, Devriendt N, Van Goethem B, Blasi F, Brioschi C, Polis I, De Cock H, Miragoli L, Oliva P, Valbusa G, Vanderperren K, de Rooster H. Fluorescence-guided surgery using indocyanine green in dogs with superficial solid tumours. Vet Rec 2020; 187:273. [PMID: 32345608 DOI: 10.1136/vr.105554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 04/01/2020] [Accepted: 04/05/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Near-infrared fluorescence (NIRF) imaging is a relatively novel technique that can aid surgeons during intraoperative tumour identification. METHODS Nine canine oncology patients (five mammary gland tumours, three mast cell tumours and one melanoma) received intravenous indocyanine green (ICG). After 24 hours, tumours were resected and fluorescence intensities of tumours and surroundings were evaluated. Additional wound bed tissue was resected if residual fluorescence was present after tumour resection. Ex vivo, fluorescence-guided dissection was performed to separate tumour from surrounding tissue. RESULTS Intraoperative NIRF-guided tumour delineation was feasible in four out of nine dogs. Wound bed imaging after tumour removal identified nine additional fluorescent lesions, of which four contained tumour tissue. One of these four true positive in vivo lesions was missed by standard-of-care inspection. Ex vivo fluorescence-guided tumour dissection showed a sensitivity of 72 per cent and a specificity of 80 per cent in discriminating between tumour and surrounding tissue. CONCLUSION The value of ICG for intraoperative tumour delineation seems more limited than originally thought. Although NIRF imaging using ICG did identify remaining tumour tissue in the wound bed, a high false positive rate was also observed.
Collapse
Affiliation(s)
- Sophie Favril
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium .,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| | - Eline Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| | - Emmelie Stock
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Nausikaa Devriendt
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bart Van Goethem
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | - Ingeborgh Polis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | - Paolo Oliva
- Bracco Imaging SpA, Colleretto Giacosa, Italy
| | | | - Katrien Vanderperren
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hilde de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| |
Collapse
|
34
|
Surface-Registration Frameless Stereotactic Navigation Is Less Accurate During Prone Surgeries: Intraoperative Near-Infrared Visualization Using Second Window Indocyanine Green Offers an Adjunct. Mol Imaging Biol 2020; 22:1572-1580. [PMID: 32232627 DOI: 10.1007/s11307-020-01495-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Frameless neuronavigation allows neurosurgeons to visualize and relate the position of surgical instruments to intracranial pathologies based on preoperative tomographic imaging. However, neuronavigation can often be inaccurate. Multiple factors have been proposed as potential causes, and new technologies are needed to overcome these challenges. OBJECTIVE To evaluate the accuracy of neuronavigation systems compared to near-infrared (NIR) fluorescence imaging using Second Window Indocyanine Green, a novel technique, and to determine factors that lead to neuronavigation errors. METHODS A retrospective analysis was conducted on 56 patients who underwent primary resections of intracranial tumors. Patients received 5 mg/kg ICG approximately 24 h preoperatively. Intraoperatively, neuronavigation was used to plan craniotomies to place the tumors in the center. After craniotomy, NIR imaging visualized tumor-specific NIR signals. The accuracy of neuronavigation and NIR fluorescence imaging for delineating the tumor boundary prior to durotomy was compared. RESULTS The neuronavigation centers and NIR centers were 23.0 ± 7.7 % and 2.6 ± 1.1 % deviated from the tumor centers, respectively, relative to the craniotomy sizes. In 12 cases, significant changes were made to the planned durotomy based on NIR imaging. Patient position was a significant predictor of neuronavigation inaccuracy on both univariate and multivariate analysis, with the prone position having significantly higher inaccuracy (29.2 ± 8.1 %) compared to the supine (16.2 ± 8.1 %, p value < 0.001) or the lateral (17.9 ± 5.1 %, p value = 0.003) positions. CONCLUSION Patient position significantly affects neuronavigation accuracy. Intraoperative NIR fluorescence imaging before durotomy offers an opportunity to readjust the neuronavigation image space to better align with the patient space.
Collapse
|
35
|
Wu T, Wang X, Zhang R, Jiao Y, Yu W, Su D, Zhao Y, Tian J. Mice with pre-existing tumors are vulnerable to postoperative cognitive dysfunction. Brain Res 2020; 1732:146650. [DOI: 10.1016/j.brainres.2020.146650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 01/01/2020] [Accepted: 01/06/2020] [Indexed: 02/07/2023]
|
36
|
Cho SS, Jeon J, Buch L, Nag S, Nasrallah M, Low PS, Grady MS, Singhal S, Lee JYK. Intraoperative near-infrared imaging with receptor-specific versus passive delivery of fluorescent agents in pituitary adenomas. J Neurosurg 2019; 131:1974-1984. [PMID: 30554181 PMCID: PMC10985533 DOI: 10.3171/2018.7.jns181642] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/31/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Intraoperative molecular imaging with tumor-targeted fluorescent dyes can enhance resection rates. In contrast to visible-light fluorophores (e.g., 5-aminolevulinic-acid), near-infrared (NIR) fluorophores have increased photon tissue penetration and less contamination from tissue autofluorescence. The second-window ICG (SWIG) technique relies on passive accumulation of indocyanine green (ICG) in neoplastic tissues. OTL38, conversely, targets folate receptor overexpression in nonfunctioning pituitary adenomas. In this study, we compare the properties of these 2 modalities for NIR imaging of pituitary adenomas to better understand the potential for NIR imaging in neurosurgery. METHODS A total of 39 patients with pituitary adenomas were enrolled between June 2015 and January 2018 in 2, sequential, IRB-approved studies. Sixteen patients received systemic ICG infusions 24 hours prior to surgery, and another 23 patients received OTL38 infusions 2-3 hours prior to surgery. NIR fluorescence signal-to-background ratio (SBR) was recorded during and after resection. Immunohistochemistry was performed on the 23 adenomas resected from patients who received OTL38 to assess expression of folate receptor-alpha (FRα). RESULTS All 16 adenomas operated on after ICG administration demonstrated strong NIR fluorescence (mean SBR 4.1 ± 0.69 [SD]). There was no statistically significant difference between the 9 functioning and 7 nonfunctioning adenomas (p = 0.9). After administration of OTL38, the mean SBR was 1.7 ± 0.47 for functioning adenomas, 2.6 ± 0.91 for all nonfunctioning adenomas, and 3.2 ± 0.53 for the subset of FRα-overexpressing adenomas. Tissue identification with white light alone for all adenomas demonstrated 88% sensitivity and 90% specificity. SWIG demonstrated 100% sensitivity but only 29% specificity for both functioning and nonfunctioning adenomas. OTL38 was 75% sensitive and 100% specific for all nonfunctioning adenomas, but when assessment was limited to the 9 FRα-overexpressing adenomas, the sensitivity and specificity of OTL38 were both 100%. CONCLUSIONS Intraoperative imaging with NIR fluorophores demonstrates highly sensitive detection of pituitary adenomas. OTL38, a folate-receptor-targeted fluorophore, is highly specific for nonfunctioning adenomas but has no utility in functioning adenomas. SWIG, which relies on passive diffusion into neoplastic tissue, is applicable to both functioning and nonfunctioning pituitary adenomas, but it is less specific than targeted fluorophores. Thus, targeted and nontargeted NIR fluorophores play important, yet distinct, roles in intraoperative imaging. Selectively and intelligently using either agent has the potential to greatly improve resection rates and outcomes for patients with intracranial tumors.
Collapse
Affiliation(s)
- Steve S. Cho
- Department of Neurosurgery Hospital of the University of Pennsylvania
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jun Jeon
- Department of Neurosurgery Hospital of the University of Pennsylvania
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Love Buch
- Department of Neurosurgery Hospital of the University of Pennsylvania
| | - Shayoni Nag
- Department of Neurosurgery Hospital of the University of Pennsylvania
| | | | - Philip S. Low
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - M. Sean Grady
- Department of Neurosurgery Hospital of the University of Pennsylvania
| | - Sunil Singhal
- Department of Surgery Hospital of the University of Pennsylvania
| | - John Y. K. Lee
- Department of Neurosurgery Hospital of the University of Pennsylvania
| |
Collapse
|
37
|
Dacy A, Haider N, Davis K, Hu W, Tang L. Design and evaluation of an imager for assessing wound inflammatory responses and bioburden in a pig model. JOURNAL OF BIOMEDICAL OPTICS 2019; 25:1-9. [PMID: 31515974 PMCID: PMC6739619 DOI: 10.1117/1.jbo.25.3.032002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Our work details the development and characterization of a portable luminescence imaging device for detecting inflammatory responses and infection in skin wounds. The device includes a CCD camera and close-up lens integrated into a customizable 3D printed imaging chamber to create a portable light-tight imager for luminescence imaging. The chamber has an adjustable light portal that permits ample ambient light for white light imaging. This imager was used to quantify in real time the extent of two-dimensional reactive oxygen species (ROS) activity distribution using a porcine wound infection model. The imager was used to successfully visualize ROS-associated luminescent activities in vitro and in vivo. Using a pig full-thickness cutaneous wound model, we further demonstrate that this portable imager can detect the change of ROS activities and their relationship with vasculature in the wound environment. Finally, by analyzing ROS intensity and distribution, an imaging method was developed to distinguish infected from uninfected wounds. We discovered a distinct ROS pattern between bacteria-infected and control wounds corresponding to the microvasculature. The results presented demonstrate that this portable luminescence imager is capable of imaging ROS activities in cutaneous wounds in a large animal model, indicating suitability for future clinical applications.
Collapse
Affiliation(s)
- Ashley Dacy
- University of Texas at Arlington, Department of Bioengineering, Arlington, Texas, United States
| | - Nowmi Haider
- University of Texas at Arlington, Department of Bioengineering, Arlington, Texas, United States
| | - Kathryn Davis
- University of Texas Southwestern Medical Center, Department of Plastic Surgery, Dallas, Texas, United States
| | - Wenjing Hu
- Progenitec Inc., Arlington, Texas, United States
| | - Liping Tang
- University of Texas at Arlington, Department of Bioengineering, Arlington, Texas, United States
| |
Collapse
|
38
|
Newton AD, Predina JD, Shin MH, Frenzel-Sulyok LG, Vollmer CM, Drebin JA, Singhal S, Lee MK. Intraoperative Near-infrared Imaging Can Identify Neoplasms and Aid in Real-time Margin Assessment During Pancreatic Resection. Ann Surg 2019; 270:12-20. [PMID: 31188797 PMCID: PMC11068217 DOI: 10.1097/sla.0000000000003201] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To determine if intraoperative near-infrared (NIR) imaging carries benefit in resection of pancreatic neoplasms. BACKGROUND Resection of pancreatic malignancies is hindered by high rates of local and distant recurrence from positive margins and unrecognized metastases. Improved tumor visualization could improve outcomes. We hypothesized that intraoperative NIR imaging with a clinically approved optical contrast agent could serve as a useful adjunct in assessing margins and extent of disease during pancreatic resections. METHODS Twenty patients were enrolled in an open-label clinical trial from July 2016 to May 2018. Subjects received second window indocyanine green (ICG) (2.5-5 mg/kg) 24 hours prior to pancreatic resection. NIR imaging was performed during staging laparoscopy and after pancreas mobilization in situ and following resection ex vivo. Tumor fluorescence was quantified using tumor-to-background ratio (TBR). Fluorescence at the specimen margin was compared to pathology evaluation. RESULTS Procedures included 9 pancreaticoduodenectomies, 10 distal pancreatectomies, and 1 total pancreatectomy; 21 total specimens were obtained. Three out of 8 noninvasive tumors were fluorescent (mean TBR 2.59 ± 2.57). Twelve out of 13 invasive malignancies (n = 12 pancreatic adenocarcinoma, n = 1 cholangiocarcinoma) were fluorescent (mean TBR 4.42 ± 2.91). Fluorescence at the transection margin correlated with final pathologic assessment in 12 of 13 patients. Following neoadjuvant therapy, 4 of 5 tumors were fluorescent; these 4 tumors showed no treatment response on pathology assessment. One tumor had a significant treatment response and showed no fluorescence. CONCLUSIONS Second window ICG reliably accumulates in invasive pancreatic malignancies and provides real-time feedback during pancreatectomy. NIR imaging may help to assess the response to neoadjuvant therapy.
Collapse
Affiliation(s)
- Andrew D. Newton
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jarrod D. Predina
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Michael H. Shin
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Lydia G. Frenzel-Sulyok
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Charles M. Vollmer
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jeffrey A. Drebin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Major K. Lee
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
39
|
Olson MT, Ly QP, Mohs AM. Fluorescence Guidance in Surgical Oncology: Challenges, Opportunities, and Translation. Mol Imaging Biol 2019; 21:200-218. [PMID: 29942988 PMCID: PMC6724738 DOI: 10.1007/s11307-018-1239-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Surgical resection continues to function as the primary treatment option for most solid tumors. However, the detection of cancerous tissue remains predominantly subjective and reliant on the expertise of the surgeon. Surgery that is guided by fluorescence imaging has shown clinical relevance as a new approach to detecting the primary tumor, tumor margins, and metastatic lymph nodes. It is a technique to reduce recurrence and increase the possibility of a curative resection. While significant progress has been made in developing this emerging technology as a tool to assist the surgeon, further improvements are still necessary. Refining imaging agents and tumor targeting strategies to be a precise and reliable surgical strategy is essential in order to translate this technology into patient care settings. This review seeks to provide a comprehensive update on the most recent progress of fluorescence-guided surgery and its translation into the clinic. By highlighting the current status and recent developments of fluorescence image-guided surgery in the field of surgical oncology, we aim to offer insight into the challenges and opportunities that require further investigation.
Collapse
Affiliation(s)
- Madeline T Olson
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Quan P Ly
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Aaron M Mohs
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 5-12315 Scott Research Tower, Omaha, NE, 68198, USA.
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
40
|
Cho SS, Salinas R, Lee JYK. Indocyanine-Green for Fluorescence-Guided Surgery of Brain Tumors: Evidence, Techniques, and Practical Experience. Front Surg 2019; 6:11. [PMID: 30915339 PMCID: PMC6422908 DOI: 10.3389/fsurg.2019.00011] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 02/19/2019] [Indexed: 01/10/2023] Open
Abstract
The primary treatment for brain tumors often involves surgical resection for diagnosis, relief of mass effect, and prolonged survival. In neurosurgery, it is of utmost importance to achieve maximal safe resection while minimizing iatrogenic neurologic deficit. Thus, neurosurgeons often rely on extra tools in the operating room, such as neuronavigation, intraoperative magnetic resonance imaging, and/or intraoperative rapid pathology. However, these tools can be expensive, not readily available, time-consuming, and/or inaccurate. Recently, fluorescence-guided surgery has emerged as a cost-effective method to accurately visualize neoplastic areas in real-time to guide resection. Currently, 5-aminolevulinic-acid (5-ALA) remains the only fluorophore that has been approved specifically for fluorescence-guided tumor resection. Its use has demonstrated improved resection rates and prolonged progression-free survival. However, protoporphyrin-IX, the metabolic product of 5-ALA that accumulates in neoplastic cells, fluoresces in the visible-light range, which suffers from limited tissue penetration and significant auto-fluorescence. Near-infrared fluorescence, on the other hand, overcomes these problems with ease. Since 2012, researchers at our institution have developed a novel technique using indocyanine-green, which is a well-known near-infrared fluorophore used traditionally for angiography. This Second-Window-ICG (SWIG) technique takes advantage of the increased endothelial permeability in peritumoral tissue, which allows indocyanine-green to accumulate in these areas for intraoperative visualization of the tumor. SWIG has demonstrated utility in gliomas, meningiomas, metastases, pituitary adenomas, chordomas, and craniopharyngiomas. The main benefits of SWIG stem from its highly sensitive detection of neoplastic tissue in a wide variety of intracranial pathologies in real-time, which can help neurosurgeons both during surgical resections and in stereotactic biopsies. In this review of this novel technique, we summarize the development and mechanism of action of SWIG, provide evidence for its benefits, and discuss its limitations. Finally, for those interested in near-infrared fluorescence-guided surgery, we provide suggestions for maximizing the benefits while minimizing the limitations of SWIG based on our own experience thus far.
Collapse
Affiliation(s)
- Steve S Cho
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Ryan Salinas
- Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - John Y K Lee
- Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
41
|
Predina JD, Okusanya O, D Newton A, Low P, Singhal S. Standardization and Optimization of Intraoperative Molecular Imaging for Identifying Primary Pulmonary Adenocarcinomas. Mol Imaging Biol 2018; 20:131-138. [PMID: 28497233 DOI: 10.1007/s11307-017-1076-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Intraoperative molecular imaging (IMI) is an emerging technology used to locate pulmonary adenocarcinomas and identify positive margins during surgery. Background noise and tissue autofluorescence have been major obstacles. The goal of this study is to optimize the image quality of folate receptor alpha (FRα) targeted IMI for pulmonary adenocarcinomas by modifying emission data. PROCEDURES A total of 15 lung cancer patients were enrolled in a pilot study. In the first cohort, FRα upregulation within pulmonary adenocarcinoma tumors was confirmed by analyzing specimens from five pulmonary adenocarcinoma patients with flow cytometry and immunohistochemistry. Next, in a cohort of five additional patients, autofluorescence of intrathoracic structures and tissues was quantified. Lastly, five patients with tumors at various depths from the pleural surface were enrolled and received the FRα-targeted optical contrast agent, EC17. In this final cohort, resected pulmonary adenocarcinomas were imaged at a wide range of fluorescence exposure times (0 to 200 ms), various laser powers, and with unique filter configurations. Tumor-to-noise ratio (TNR) for images was generated using region of interest software. RESULTS Pulmonary adenocarcinomas highly express FRα. Significant autofluorescence from native thoracic tissues was found with the highest fluorescent signals at the bronchial stump (547 ± 98, range 423-699), the pulmonary artery (267 ± 64, range 200-374), and cortical bone (266 ± 17, range 243-287). High levels of autofluorescence were appreciated after systemic administration of EC17; however, TNR was improved by altering exposure settings at the time of the imaging. Optimal fluorescent exposure time occurs at 40 ms (25 frames/s). CONCLUSIONS Exposure properties can be manipulated to maximize TNR thus allowing for successful intraoperative detection of pulmonary adenocarcinomas during surgery. Optimization of the conditions for intraoperative molecular imaging sets the stage for future clinical trials utilizing targeted IMI techniques which can aid the surgeon at the time of cancer resection.
Collapse
Affiliation(s)
- Jarrod D Predina
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Olugbenga Okusanya
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Andrew D Newton
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Philip Low
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Sunil Singhal
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA. .,Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
42
|
Newton AD, Predina JD, Corbett CJ, Frenzel-Sulyok LG, Xia L, Petersson EJ, Tsourkas A, Nie S, Delikatny EJ, Singhal S. Optimization of Second Window Indocyanine Green for Intraoperative Near-Infrared Imaging of Thoracic Malignancy. J Am Coll Surg 2018; 228:188-197. [PMID: 30471345 DOI: 10.1016/j.jamcollsurg.2018.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Near-infrared (NIR) imaging using the second time window of indocyanine green (ICG) allows localization of pulmonary, pleural, and mediastinal malignancies during surgery. Based on empirical evidence, we hypothesized that different histologic tumor types fluoresce optimally at different ICG doses. STUDY DESIGN Patients with thoracic tumors biopsy-proven or suspicious for malignancy were enrolled in an NIR imaging clinical trial. Patients received a range of ICG doses 1 day before surgery: 1 mg/kg (n = 8), 2 mg/kg (n = 8), 3 mg/kg (n = 13), 4 mg/kg (n = 8), and 5 mg/kg (n = 8). Intraoperatively, NIR imaging was performed. The endpoint was to identify the highest tumor-to-background fluorescence ratio (TBR) for each tumor type at each dose. Final pathology confirmed tumor histology. RESULTS Of 45 patients, 41 had malignancies (18 non-small cell lung cancers [NSCLC], 3 pulmonary neuroendocrine tumors, 13 thoracic metastases, 4 thymomas, 3 mesotheliomas). At doses of 4 to 5 mg/kg, the TBR from primary NSCLC vs other malignancies was no different (2.70 vs 3.21, p = 1.00). At doses of 1 to 3 mg/kg, the TBR was greater for the NSCLCs (3.19 vs 1.49, p = 0.0006). Background fluorescence from the heart or ribs was observed in 1 of 16 cases at 1 to 2 mg/kg, 5 of 13 cases at 3 mg/kg, and 14 of 16 cases at 4 to 5 mg/kg; this was a major determinant of dose optimization. CONCLUSIONS This is the first study to demonstrate that the optimal NIR contrast agent dose varies by tumor histology. Lower dose ICG (2 to 3 mg/kg) is superior for nonprimary lung cancers, and high dose ICG (4 to 5 mg/kg) is superior for lung cancers. This will have major implications as more intraoperative imaging trials surface in other specialties, will significantly reduce costs and may facilitate wider application.
Collapse
Affiliation(s)
- Andrew D Newton
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.
| | - Jarrod D Predina
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Christopher J Corbett
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Lydia G Frenzel-Sulyok
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Leilei Xia
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - E James Petersson
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Shuming Nie
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Edward J Delikatny
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
43
|
Tummers WS, Warram JM, van den Berg NS, Miller SE, Swijnenburg RJ, Vahrmeijer AL, Rosenthal EL. Recommendations for reporting on emerging optical imaging agents to promote clinical approval. Am J Cancer Res 2018; 8:5336-5347. [PMID: 30555550 PMCID: PMC6276089 DOI: 10.7150/thno.27384] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/22/2018] [Indexed: 01/12/2023] Open
Abstract
Intraoperative fluorescence imaging is particularly well-suited for surgical applications due to its inherently high sensitivity, resolution, and ability to provide images in real-time. To date, the intraoperative observation of fluorescence has largely been subjective. With the need to show objective evidence in order to demonstrate the benefit of this technique, quantitative data needs to be provided to overseeing regulatory bodies. Standardization of fluorescence imaging protocols would improve reproducibility and minimize inter- and intra-institution variance. This would allow studies to be conducted using the same injection techniques, imaging times, reconstruction methods, and analyses. Here, we provide recommendations for standardized methodologies with the goal of setting a minimum requirement for reporting fluorescence-guided surgery results based on both qualitative and (semi-) quantitative data collection. Clinical trials using fluorescence-guided surgery should present results of three critical elements; 1) intra-operative imaging, 2) specimen mapping and pathology correlation, and 3) target validation. Qualitative analyses should consist of a bright field image, black-and-white fluorescence image, pseudo-colored fluorescence overlay image, and/or heat-map whereby fluorescence signal intensity differences are displayed on a color spectrum. Quantitative analyses should include 1) intraoperative data (consisting of images or video, raw numeric values and ratios); 2) specimen mapping, for correlation of fluorescence with the presence of disease (performed using fresh tissue); and 3) target validation (designed to determine fluorescence intensity relative to receptor density of a specific area). Including the aforementioned methods of both qualitative and quantitative analyses will ensure that trial results are comparable and could be collated in future studies to expedite FDA approval.
Collapse
|
44
|
Peng L, Shang W, Guo P, He K, Wang H, Han Z, Jiang H, Tian J, Wang K, Xu W. Phage Display-Derived Peptide-Based Dual-Modality Imaging Probe for Bladder Cancer Diagnosis and Resection Postinstillation: A Preclinical Study. Mol Cancer Ther 2018; 17:2100-2111. [PMID: 30082470 DOI: 10.1158/1535-7163.mct-18-0212] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/09/2018] [Accepted: 08/02/2018] [Indexed: 11/16/2022]
Abstract
Bladder cancer is a common human malignancy. Conventional ultrasound and white-light cystoscopy are often used for bladder cancer diagnosis and resection, but insufficient specificity results in a high bladder cancer recurrence rate. New strategies for the diagnosis and resection of bladder cancer are needed. In this study, we developed a highly specific peptide-based probe for bladder cancer photoacoustic imaging (PAI) diagnosis and near-infrared (NIR)-imaging-guided resection after instillation. A bladder cancer-specific peptide (PLSWT7) was selected by in vivo phage-display technology and labeled with IRDye800CW to synthesize a bladder cancer-specific dual-modality imaging (DMI) probe (PLSWT7-DMI). The feasibility of PLSWT7-DMI-based dual-modality PAI-NIR imaging was assessed in vitro, in mouse models, and ex vivo human bladders. An air-pouch bladder cancer (APBC) model suitable for probe instillation was established to evaluate the probe-based bladder cancer PAI diagnosis and NIR-imaging-guided resection. Human bladders were used to assess whether the PLSWT7-DMI-based DMI strategy is a translatable approach for bladder cancer detection and resection. The probe exhibited excellent selectivity and specificity both in vitro and in vivo Postinstillation of the probe, tumors <3 mm were detectable by PAI, and NIR-imaging-guided tumor resection decreased the bladder cancer recurrence rate by 90% and increased the survival in the mouse model. Additionally, ex vivo NIR imaging of human bladders indicated that PLSWT7-DMI-based imaging would potentially allow precise resection of bladder cancer in clinical settings. This PLSWT7-DMI-based DMI strategy was a translatable approach for bladder cancer diagnosis and resection and could potentially lower the bladder cancer recurrence rate. Mol Cancer Ther; 17(10); 2100-11. ©2018 AACR.
Collapse
Affiliation(s)
- Li Peng
- Urology Surgery Department, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P.R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, Heilongjiang, P.R. China
| | - Wenting Shang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P.R. China
- The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Pengyu Guo
- Urology Surgery Department, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P.R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, Heilongjiang, P.R. China
| | - Kunshan He
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P.R. China
- The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Hongzhi Wang
- Urology Surgery Department, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, Heilongjiang, P.R. China
| | - Ziyu Han
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P.R. China
- The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Hongmei Jiang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P.R. China
- The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P.R. China.
- The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Beihang University, Beijing, P.R. China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, P.R. China.
- The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Wanhai Xu
- Urology Surgery Department, The Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China.
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, Heilongjiang, P.R. China
| |
Collapse
|
45
|
Newton AD, Predina JD, Nie S, Low PS, Singhal S. Intraoperative fluorescence imaging in thoracic surgery. J Surg Oncol 2018; 118:344-355. [PMID: 30098293 DOI: 10.1002/jso.25149] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/04/2018] [Indexed: 12/19/2022]
Abstract
Intraoperative fluorescence imaging (IFI) can improve real-time identification of cancer cells during an operation. Phase I clinical trials in thoracic surgery have demonstrated that IFI with second window indocyanine green (TumorGlow® ) can identify subcentimeter pulmonary nodules, anterior mediastinal masses, and mesothelioma, while the use of a folate receptor-targeted near-infrared agent, OTL38, can improve the specificity for diagnosing tumors with folate receptor expression. Here, we review the existing preclinical and clinical data on IFI in thoracic surgery.
Collapse
Affiliation(s)
- Andrew D Newton
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jarrod D Predina
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shuming Nie
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
46
|
Lee JYK, Cho SS, Zeh R, Pierce JT, Martinez-Lage M, Adappa ND, Palmer JN, Newman JG, Learned KO, White C, Kharlip J, Snyder P, Low PS, Singhal S, Grady MS. Folate receptor overexpression can be visualized in real time during pituitary adenoma endoscopic transsphenoidal surgery with near-infrared imaging. J Neurosurg 2018; 129:390-403. [PMID: 28841122 PMCID: PMC10980838 DOI: 10.3171/2017.2.jns163191] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Pituitary adenomas account for approximately 10% of intracranial tumors and have an estimated prevalence of 15%-20% in the general US population. Resection is the primary treatment for pituitary adenomas, and the transsphenoidal approach remains the most common. The greatest challenge with pituitary adenomas is that 20% of patients develop tumor recurrence. Current approaches to reduce recurrence, such as intraoperative MRI, are costly, associated with high false-positive rates, and not recommended. Pituitary adenomas are known to overexpress folate receptor alpha (FRα), and it was hypothesized that OTL38, a folate analog conjugated to a near-infrared (NIR) fluorescent dye, could provide real-time intraoperative visual contrast of the tumor versus the surrounding nonneoplastic tissues. The preliminary results of this novel clinical trial are presented. METHODS Nineteen adult patients who presented with pituitary adenoma were enrolled. Patients were infused with OTL38 2-4 hours prior to surgery. A 4-mm endoscope with both visible and NIR light capabilities was used to visualize the pituitary adenoma and its margins in real time during surgery. The signal-to-background ratio (SBR) was recorded for each tumor and surrounding tissues at various endoscope-to-sella distances. Immunohistochemical analysis was performed to assess the FRα expression levels in all specimens and classify patients as having either high or low FRα expression. RESULTS Data from 15 patients (4 with null cell adenomas, 1 clinically silent gonadotroph, 1 totally silent somatotroph, 5 with a corticotroph, 3 with somatotrophs, and 1 somatocorticotroph) were analyzed in this preliminary analysis. Four patients were excluded for technical considerations. Intraoperative NIR imaging delineated the main tumors in all 15 patients with an average SBR of 1.9 ± 0.70. The FRα expression level of the adenomas and endoscope-to-sella distance had statistically significant impacts on the fluorescent SBRs. Additional considerations included adenoma functional status and time from OTL38 injection. SBRs were 3.0 ± 0.29 for tumors with high FRα expression (n = 3) and 1.6 ± 0.43 for tumors with low FRα expression (n = 12; p < 0.05). In 3 patients with immunohistochemistry-confirmed FRα overexpression (2 patients with null cell adenoma and 1 patient with clinically silent gonadotroph), intraoperative NIR imaging demonstrated perfect classification of the tumor margins with 100% sensitivity and 100% specificity. In addition, for these 3 patients, intraoperative residual fluorescence predicted postoperative MRI results with perfect concordance. CONCLUSIONS Pituitary adenomas and their margins can be intraoperatively visualized with the preoperative injection of OTL38, a folate analog conjugated to NIR dye. Tumor-to-background contrast is most pronounced in adenomas that overexpress FRα. Intraoperative SBR at the appropriate endoscope-to-sella distance can predict adenoma FRα expression status in real time. This work suggests that for adenomas with high FRα expression, it may be possible to identify margins and to predict postoperative MRI findings.
Collapse
Affiliation(s)
- John Y. K. Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steve S. Cho
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ryan Zeh
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - John T. Pierce
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria Martinez-Lage
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nithin D. Adappa
- Department of Otorhinolaryngology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - James N. Palmer
- Department of Otorhinolaryngology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason G. Newman
- Department of Otorhinolaryngology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kim O. Learned
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Caitlin White
- Department of Endocrinology Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Julia Kharlip
- Department of Endocrinology Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter Snyder
- Department of Endocrinology Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Philip S. Low
- Department of Biochemistry, Purdue University, West Lafayette, Indiana
| | - Sunil Singhal
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - M. Sean Grady
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
47
|
Xiao Q, Chen T, Chen S. Fluorescent contrast agents for tumor surgery. Exp Ther Med 2018; 16:1577-1585. [PMID: 30186374 PMCID: PMC6122374 DOI: 10.3892/etm.2018.6401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 04/13/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of cases of mortality worldwide. The most effective method to cure solid tumors is surgery. Every year, >50% of cancer patients receive surgery to remove solid tumors. Surgery may increase the cure rate of most solid tumors by 4–11 fold. Surgery has many challenges, including identifying small lesions, locating metastases and confirming complete tumor removal. Fluorescence guidance describes a new approach to improve surgical accuracy. Near-infrared fluorescence imaging allows for real-time early diagnosis and intraoperative imaging of lesion tissue. The results of previous preclinical studies in the field of near-infrared fluorescence imaging are promising. This review provides examples introducing the three kinds of fluorescent dyes: The passive fluorescent dye indocyanine green, which has been approved by the Food and Drug Administration for clinical use in the USA, the fluorescent prodrug 5-aminolevulinic acid, a porphyrin precursor in the heme synthesis, and biomarker-targeted fluorescent dyes, which allow conjugation to different target sites.
Collapse
Affiliation(s)
- Qi Xiao
- School of Life Science, Nanjing Normal University, Nanjing, Jiangsu 210046, P.R. China
| | - Tianming Chen
- Department of Surgery, Nanjing Medical University Third Affiliated Hospital, Nanjing, Jiangsu 211166, P.R. China
| | - Shilin Chen
- Department of Thoracic Surgery, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
48
|
Newton AD, Predina JD, Frenzel-Sulyok LG, Shin MH, Wang Y, Singhal S. Intraoperative near-infrared imaging can identify sub-centimeter colorectal cancer lung metastases during pulmonary metastasectomy. J Thorac Dis 2018; 10:E544-E548. [PMID: 30174930 DOI: 10.21037/jtd.2018.06.161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Andrew D Newton
- Center for Precision Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jarrod D Predina
- Center for Precision Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lydia G Frenzel-Sulyok
- Center for Precision Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael H Shin
- Center for Precision Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yiqing Wang
- College of Engineering and Applied Sciences, Nanjing University, Nanjing 210000, China
| | - Sunil Singhal
- Center for Precision Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
49
|
Wang Z, Ni K, Zhang X, Ai S, Guan W, Cai H, Wang Y, Lu Q, Lane LA. Method for Real-Time Tissue Quantification of Indocyanine Green Revealing Optimal Conditions for Near Infrared Fluorescence Guided Surgery. Anal Chem 2018; 90:7922-7929. [PMID: 29864280 DOI: 10.1021/acs.analchem.8b00480] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Near infrared fluorescence guided surgery (NIRFGS) offers better distinction between cancerous and normal tissues compared to surgeries relying on a surgeon's senses of sight and touch. Because of the greater accuracy in determining tumor tissue margins, NIRFGS within clinics continues to grow. However, NIRFGS lacks standardization of the indocyanine green (ICG) dose and the preoperative period allowed after ICG administration. In an aim to find optimal doses and preoperative periods for NIRFGS standardization, we developed a method that quantitatively determines ICG levels within tissues in real-time. We find that not only do the dose and the preoperative periods influence tumor-to-background ratios (TBRs), but both also heavily influence subject-to-subject variances of these ratios. Optimal detection conditions are observed when larger than typical ICG doses are administered and longer than typical preoperative periods are allowed. Larger doses lead to increased TBRs, but longer preoperative periods are necessary to reduce TBR variances to those observed when using smaller doses. Our results suggest that a clinical investigation into maximum tolerable ICG doses and prolonging preoperative periods in NIRFGS is warranted.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China
| | - Kena Ni
- Department of Biomedical Engineering, College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China
| | - Xudong Zhang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China
| | - Shichao Ai
- Department of General Surgery, Drum Tower Hospital , Medical School of Nanjing University , Nanjing , Jiangsu 210008 , China
| | - Wenxian Guan
- Department of General Surgery, Drum Tower Hospital , Medical School of Nanjing University , Nanjing , Jiangsu 210008 , China
| | - Huiming Cai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China
| | - Yiqing Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China
| | - Qian Lu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China
| | - Lucas A Lane
- Department of Biomedical Engineering, College of Engineering and Applied Sciences , Nanjing University , Nanjing , Jiangsu 210093 , China
| |
Collapse
|
50
|
Nagahara R, Onda N, Yamashita S, Kojima M, Inohana M, Eguchi A, Nakamura M, Matsumoto S, Yoshida T, Shibutani M. Fluorescence tumor imaging by i.v. administered indocyanine green in a mouse model of colitis-associated colon cancer. Cancer Sci 2018. [PMID: 29520973 PMCID: PMC5980401 DOI: 10.1111/cas.13564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fluorescence tumor imaging using exogenous fluorescent tumor‐targeting agents has potential to improve early tumor detection. The fluorescent contrast agent indocyanine green (ICG) is used in medical diagnostics. The aim of the present study is to investigate the tumor imaging capability and the imaging mechanism of i.v. administered ICG in a mouse model of colitis‐associated colon cancer. To do this, an azoxymethane/dextran sodium sulfate‐induced colon cancer mouse model was used. Ex vivo imaging experiments were carried out 1 hour after i.v. injection of ICG. The ICG fluorescence was observed in the colon tumor tissues, with sufficient tumor to normal tissue ratio, correlating with tumor malignancy. In the tumor tissues, ICG fluorescence was localized in the vascular interstitial tissue. Immunofluorescence microscopy revealed that tumor cells formed tight junctions normally, suggesting an inability of tumor cellular uptake of ICG. In contrast, tumor tissues increased the CD31‐immunoreactive endothelial cell area, and accumulated stromal cells immunoreactive for COX‐2 and tumor cell population immunoreactive for inducible nitric oxide synthase. In vivo vascular permeability assay revealed that prostaglandin E2 promoted the endothelial cell permeability of ICG. In conclusion, our data indicated that fluorescence contrast‐enhanced imaging following i.v. administered ICG can be applied to the detection of colon tumors in a mouse colitis‐associated colon cancer model. The tumor tissue preference of ICG in the present model can be attributed to the enhanced vascular leakage of ICG involving inflammatory mediators, such as COX‐2 and inducible nitric oxide synthase, in conjunction with increased tumor vascularity.
Collapse
Affiliation(s)
- Rei Nagahara
- Division of Animal Life Science, Laboratory of Veterinary Pathology, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Nobuhiko Onda
- Division of Animal Life Science, Laboratory of Veterinary Pathology, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan.,Evaluation Technology Department 1, R&D Group, Olympus Corporation, Tokyo, Japan
| | - Susumu Yamashita
- Evaluation Technology Department 1, R&D Group, Olympus Corporation, Tokyo, Japan
| | - Miho Kojima
- Division of Animal Life Science, Laboratory of Veterinary Pathology, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan.,Evaluation Technology Department 1, R&D Group, Olympus Corporation, Tokyo, Japan
| | - Mari Inohana
- Division of Animal Life Science, Laboratory of Veterinary Pathology, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ayumi Eguchi
- Division of Animal Life Science, Laboratory of Veterinary Pathology, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Misato Nakamura
- Division of Animal Life Science, Laboratory of Veterinary Pathology, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Shinya Matsumoto
- Evaluation Technology Department 1, R&D Group, Olympus Corporation, Tokyo, Japan
| | - Toshinori Yoshida
- Division of Animal Life Science, Laboratory of Veterinary Pathology, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Makoto Shibutani
- Division of Animal Life Science, Laboratory of Veterinary Pathology, Institute of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan.,Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|