1
|
Zhi D, Zhang X, Wan S, Chen S, Xia C, Sun G, Pu Y. Intraoperative ICG NIF Imaging Defines Surgical Margin in MRONJ: A Prospective Observational Study. Oral Dis 2025. [PMID: 40364553 DOI: 10.1111/odi.15384] [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: 01/30/2025] [Revised: 03/26/2025] [Accepted: 05/04/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND This study explored the feasibility and effectiveness of indocyanine green (ICG)-based near-infrared fluorescence (NIF) imaging for localizing bone lesions associated with medication-related osteonecrosis of the jaw (MRONJ) during surgical intervention. METHODS A prospective observational study was conducted on 68 patients who underwent surgical treatment for MRONJ at a single institution between January 2021 and April 2024. Among them, 14 patients received surgery guided by NIF imaging, while the remaining patients underwent surgery without NIF guidance. Postoperative follow-up was performed to assess clinical outcomes. RESULTS There were no significant differences between the two groups regarding age, medication type and duration, underlying disease, lesion location, clinical stage, concomitant symptoms, or surgical approach. NIF imaging remained stable in 14 patients who underwent MRONJ fluorescence surgery. The recurrence rate was lower in the NIF group (14.29%) than in the non-NIF group (27.78%). The difference in the recurrence-free survival curves between the two groups was statistically significant (p = 0.043). CONCLUSION ICG-mediated NIF imaging provides a reliable method for identifying MRONJ-related bone lesions and offers valuable intraoperative guidance, potentially improving surgical outcomes.
Collapse
Affiliation(s)
- Dashuang Zhi
- Research Institute of Stomatology, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Head and Neck Oncology Surgery, Nanjing University, Nanjing, China
| | - Xinyi Zhang
- Research Institute of Stomatology, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Head and Neck Oncology Surgery, Nanjing University, Nanjing, China
| | - Shuyun Wan
- Research Institute of Stomatology, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Trauma Orthognathic Plastic Surgery, Nanjing University, Nanjing, China
| | - Sheng Chen
- Research Institute of Stomatology, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Department of Pathology, Nanjing University, Nanjing, China
| | - Chengwan Xia
- Research Institute of Stomatology, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Trauma Orthognathic Plastic Surgery, Nanjing University, Nanjing, China
| | - Guowen Sun
- Research Institute of Stomatology, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Head and Neck Oncology Surgery, Nanjing University, Nanjing, China
| | - Yumei Pu
- Research Institute of Stomatology, Affiliated Hospital of Medical School, Nanjing Stomatological Hospital, Department of Oral and Maxillofacial Head and Neck Oncology Surgery, Nanjing University, Nanjing, China
| |
Collapse
|
2
|
Qu Q, Nie H, Hou S, Guo X, Wang F, Yang H, Chen S, Deng P, Hu Z, Tian J. Fluorescence imaging assisted precise assessment of the depth of myometrial invasion in endometrial cancer lesions. Clin Transl Med 2025; 15:e70309. [PMID: 40329855 PMCID: PMC12056491 DOI: 10.1002/ctm2.70309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Affiliation(s)
- Qiaojun Qu
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingInstitute of AutomationChinese Academy of SciencesBeijingChina
| | - Huilong Nie
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Shuang Hou
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Xiaoyong Guo
- Key Laboratory of Carcinogenesis and Translational ResearchDepartment of Gastrointestinal Cancer CenterWard I, Peking University Cancer Hospital & InstituteBeijingChina
| | - Feng Wang
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Hua Yang
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Shangqiu Chen
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Panxia Deng
- Department of GynecologyThe Fifth Affiliated Hospital of Sun Yat‐sen UniversityZhuhaiChina
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingInstitute of AutomationChinese Academy of SciencesBeijingChina
- School of Artificial IntelligenceUniversity of Chinese Academy of SciencesBeijingChina
- National Key Laboratory of Kidney DiseasesBeijingChina
| | - Jie Tian
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingInstitute of AutomationChinese Academy of SciencesBeijingChina
- School of Artificial IntelligenceUniversity of Chinese Academy of SciencesBeijingChina
- National Key Laboratory of Kidney DiseasesBeijingChina
- Key Laboratory of Big Data‐Based Precision Medicine of Ministry of Industry and Information TechnologySchool of Engineering MedicineBeihang UniversityBeijingChina
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of EducationSchool of Life Science and TechnologyXidian UniversityXi'anChina
| |
Collapse
|
3
|
Lazzini G, Gaeta R, Pollina LE, Comandatore A, Furbetta N, Morelli L, D'Acunto M. Raman spectroscopy based diagnosis of pancreatic ductal adenocarcinoma. Sci Rep 2025; 15:13240. [PMID: 40247119 PMCID: PMC12006465 DOI: 10.1038/s41598-025-98122-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Pancreatic ductal adenocarcinoma is currently the 12th most frequent form of cancer worldwide, characterized by a very low 5-year survival rate. Although several therapeutic approaches have been proposed to treat this form of pancreatic cancer, surgical resection is still commonly recognized as the most effective technique to slow down the disease progression and maximize the 5-year survival rate. Analogously, one critical issue is the ability of current diagnostic methodologies to distinguish between irregular growth of the tumor mass and surrounding inflammatory tissues. In this pilot study, we apply Raman spectroscopy, supported by a series of machine learning techniques, to distinguish among healthy, pancreatitis and ductal adenocarcinoma tissues, respectively, for a total of 15 cases. Raman spectroscopy is a label-free, non-destructive spectral technique exploiting Raman scattering. In turn, by applying a combination of principal component analysis and random forest classifier on the Raman spectral dataset, we achieved a maximum accuracy of up to ∼ 96%. Our findings clearly indicate that Raman spectroscopy could become a powerful spectral technique to support pathologists in improving pancreatic cancer diagnosis.
Collapse
Affiliation(s)
- Gianmarco Lazzini
- CNR-IBF, Istituto di Biofisica Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124, Pisa, Italy
| | - Raffele Gaeta
- Second Division of Surgical Pathology, University Hospital of Pisa, Pisa, Italy
| | | | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Niccolò Furbetta
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Luca Morelli
- Department of Surgery, Amsterdam UMC, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Mario D'Acunto
- CNR-IBF, Istituto di Biofisica Consiglio Nazionale delle Ricerche, via Moruzzi 1, 56124, Pisa, Italy.
| |
Collapse
|
4
|
Piper TB, Schaebel GH, Egeland C, Achiam MP, Burgdorf SK, Nerup N. Fluorescence-guided pancreatic surgery: A scoping review. Surgery 2025; 178:108931. [PMID: 39613658 DOI: 10.1016/j.surg.2024.10.022] [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: 04/07/2024] [Revised: 09/05/2024] [Accepted: 10/15/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Although fluorescence guidance during various surgical procedures has been shown to be safe and have possible better clinical outcomes than without the guidance, the use of fluorophores in pancreatic surgery is novel and not yet well described. This scoping review involved a systematic methodology of the currently available literature and aimed to illuminate the use of fluorophores in pancreatic surgery from a clinical view. METHODS The PRISMA and the PRISMA-ScR guidelines were used when appropriate and the following databases were searched: PubMed, Embase, Scopus, The Cochrane Collection, and Web of Science. Human original articles and case reports were included. Bias was assessed with the Newcastle-Ottawa Scale and the IDEAL framework was used for evaluation of surgical innovation. RESULTS A total of 5,565 search hits were screened, and 23 original articles and 24 case reports consisting of 754 patients met the inclusion criteria. The use of indocyanine green was both the most prominent and the most promising method for securing sufficient perfusion of neighboring organs, enhancing the detection and distinguishing of neuroendocrine tumors, and assisting in the identification of hepatic micrometastases. CONCLUSION The included studies were generally heterogenic, exploratory, and small. Indocyanine green was used in several ways, and it may add clinical value in different settings during pancreatic surgery. Tumor-targeted probes are a rapidly developing and promising field of research.
Collapse
Affiliation(s)
- Thomas B Piper
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Gustav H Schaebel
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte Egeland
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark. https://www.twitter.com/ChEgeland
| | - Michael P Achiam
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark. https://www.twitter.com/MichaelAchiam
| | - Stefan K Burgdorf
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nikolaj Nerup
- Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark. https://www.twitter.com/nikolajnerup
| |
Collapse
|
5
|
Cao Z, Hu Z, Wang Y, Zhao F, Li J, Li R, Hu W, Liu B, Tang S, Wang P, Li F, Chen Z, Wang L, Ma X. Letrozole-Based Near-Infrared Dynamic Imaging Targeting Ductal-Vascular RhoJ From Pancreatic Intraepithelial Neoplasia to Pancreatic Ductal Adenocarcinoma. Adv Healthc Mater 2024; 13:e2402913. [PMID: 39329457 DOI: 10.1002/adhm.202402913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Indexed: 09/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) relies heavily on neoangiogenesis for its progression, making early detection crucial. Here, LTZi-MHI148 (Letrozole inhibitor bonding with MHI-148 dye), a near-infrared (NIR) fluorescent agent is developed, to target RhoJ (Ras Homolog Family Member J), a protein expressed in neonatal vasculature, for both imaging and therapy of early PDAC. This agent is synthesized by conjugating Letrozole with MHI-148, exhibiting excellent NIR characteristics and photostability. In vitro studies showed that LTZi-MHI148 selectively accumulated within pancreatic cancer cells through Organic Anion Transporting Polypeptide (OATP) transporters and bound to cytoplasmic RhoJ. In vivo, the probe effectively targeted neoangiogenesis and Pancreatic Intraepithelial Neoplasias (PanINs) in various PDAC models, including the orthotopic, ectopic, spontaneous, and tamoxifen-induced tumors. Notably, LTZi-MHI148 detected preneoplastic PanIN lesions with Overexpressed RhoJ and active neoangiogenesis in both spontaneous and tamoxifen-induced PDAC murine models. Longitudinal imaging studies revealed that RhoJ-targeted neoangiogenesis tracks lesion progression, highlighting LTZi-MHI148's utility in monitoring disease progression. Furthermore, multiple LTZi-MHI148 administrations attenuated PanINs to PDAC progression, suggesting its potential as a therapeutic intervention. These findings underscore the translational potential of LTZi-MHI148 for the early detection and targeted therapy of PDAC, utilizing NIR-I/II imaging to monitor RhoJ overexpression in precancerous ductal neoplasia associated with neoangiogenesis.
Collapse
Affiliation(s)
- Zhi Cao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong, 510631, China
| | - Zhuang Hu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
| | - Yishu Wang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong, 510631, China
| | - Fengyun Zhao
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Jingmin Li
- Internal medicine department, Guangzhou women and children's Medical Center, Guangzhou, Guangdong, 510620, China
| | - Ruihan Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
| | - Weibin Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong, 510631, China
| | - Bei Liu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong, 510631, China
| | - Shaohui Tang
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Ping Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan, Guangdong, 528403, China
| | - Zilin Chen
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
| | - Lisheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, China Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong, 510631, China
| |
Collapse
|
6
|
Chen K, Teng X, Zhou N, Cheng W. Rising sun or strangled in the cradle? A narrative review of near-infrared fluorescence imaging-guided surgery for pancreatic tumors. Int J Surg 2024; 110:7929-7947. [PMID: 38768476 PMCID: PMC11634182 DOI: 10.1097/js9.0000000000001676] [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: 11/26/2023] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Near-infrared fluorescence (NIRF)-guided surgical navigation has become a promising and effective detection method in pancreatic tumor surgery. The imaging technique has gradually transitioned from the NIR-I region to the NIR-II region. Real-time assessment of the tumor boundary and determination of the ideal resection plane are essential for preserving the pancreatic parenchyma and its secretory functions. However, since the pancreatic parenchyma has a less rich blood supply than the liver, the application of contrast agents in pancreatic tumor surgery is still in its infancy. The application of indocyanine green (ICG) and methylene blue (MB) in intraoperative NIRF imaging of pancreatic tumors has become more mature, but due to the characteristics of nonspecific imaging, the imaging efficiency and depth need to be improved. Many tumor-specific imaging agents have been designed, but most of them have not gone past animal trials because of their high development and imaging costs, biotoxicity, and other limitations. In this article, we review recent reports of ICG, MB, and newly developed contrast agents and imaging devices. We focus on the current status and new developments in the application of these contrast agents and summarize the current clinical and preclinical studies on specific contrast agents. We synthesize relevant reports to discuss the difficulties and prospects of the application of fluorescent imaging agents in pancreatic tumors. We hope that reviewing previous studies and the current progress on contrast imaging technology will provide new perspectives for its future application and development in pancreatic tumor surgery, which should translate into better patient prognoses. The manuscript was written according to the Scale for the Assessment of Narrative Review Articles (SANRA).
Collapse
Affiliation(s)
- Kang Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha
| | - Xiong Teng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Ning Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha
| |
Collapse
|
7
|
Zajac J, Liu A, Hassan S, Gibson A. Mechanisms of delayed indocyanine green fluorescence and applications to clinical disease processes. Surgery 2024; 176:386-395. [PMID: 38749795 PMCID: PMC11246809 DOI: 10.1016/j.surg.2024.03.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Delayed indocyanine green fluorescence imaging is under investigation in various clinical disease processes. Understanding the mechanisms of indocyanine green accumulation and retention is essential to correctly interpreting and analyzing imaging data. The purpose of this scoping review was to synthesize what is known about the mechanism of indocyanine green retention at the cellular level to better understand the clinical nuances of delayed indocyanine green imaging and identify critical gaps in our knowledge to guide future studies. METHODS We performed a scoping review of 7,087 citations after performing database searches of PubMed, Scopus, the Cochrane Library, and the Web of Science Core Collection electronic databases. Studies were eligible for inclusion if they were peer-reviewed original research discussing the mechanism of indocyanine green retention in the results section in disease processes involving inflammation and/or necrosis, including cancer, and were available in English. Data were extracted using Covidence software. RESULTS Eighty-nine studies were included in the final analysis. Several features of indocyanine green retention were identified. CONCLUSION We identified several mechanistic features involved in indocyanine green accumulation in diseased tissue that overall had distinct mechanisms of indocyanine green retention in tumors, nontumor inflammation, and necrosis. Our study also reveals new insights on how inflammatory infiltrate influences indocyanine green fluorescence imaging. These findings are noteworthy because they add to our understanding of how fluorescence-guided surgery may be optimized based on the pathology of interest via specific indocyanine green dosing and timing of image acquisition.
Collapse
Affiliation(s)
- Jocelyn Zajac
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Aiping Liu
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Sameeha Hassan
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Angela Gibson
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
8
|
Boland PA, Hardy NP, Moynihan A, McEntee PD, Loo C, Fenlon H, Cahill RA. Intraoperative near infrared functional imaging of rectal cancer using artificial intelligence methods - now and near future state of the art. Eur J Nucl Med Mol Imaging 2024; 51:3135-3148. [PMID: 38858280 PMCID: PMC11300525 DOI: 10.1007/s00259-024-06731-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/15/2024] [Indexed: 06/12/2024]
Abstract
Colorectal cancer remains a major cause of cancer death and morbidity worldwide. Surgery is a major treatment modality for primary and, increasingly, secondary curative therapy. However, with more patients being diagnosed with early stage and premalignant disease manifesting as large polyps, greater accuracy in diagnostic and therapeutic precision is needed right from the time of first endoscopic encounter. Rapid advancements in the field of artificial intelligence (AI), coupled with widespread availability of near infrared imaging (currently based around indocyanine green (ICG)) can enable colonoscopic tissue classification and prognostic stratification for significant polyps, in a similar manner to contemporary dynamic radiological perfusion imaging but with the advantage of being able to do so directly within interventional procedural time frames. It can provide an explainable method for immediate digital biopsies that could guide or even replace traditional forceps biopsies and provide guidance re margins (both areas where current practice is only approximately 80% accurate prior to definitive excision). Here, we discuss the concept and practice of AI enhanced ICG perfusion analysis for rectal cancer surgery while highlighting recent and essential near-future advancements. These include breakthrough developments in computer vision and time series analysis that allow for real-time quantification and classification of fluorescent perfusion signals of rectal cancer tissue intraoperatively that accurately distinguish between normal, benign, and malignant tissues in situ endoscopically, which are now undergoing international prospective validation (the Horizon Europe CLASSICA study). Next stage advancements may include detailed digital characterisation of small rectal malignancy based on intraoperative assessment of specific intratumoral fluorescent signal pattern. This could include T staging and intratumoral molecular process profiling (e.g. regarding angiogenesis, differentiation, inflammatory component, and tumour to stroma ratio) with the potential to accurately predict the microscopic local response to nonsurgical treatment enabling personalised therapy via decision support tools. Such advancements are also applicable to the next generation fluorophores and imaging agents currently emerging from clinical trials. In addition, by providing an understandable, applicable method for detailed tissue characterisation visually, such technology paves the way for acceptance of other AI methodology during surgery including, potentially, deep learning methods based on whole screen/video detailing.
Collapse
Affiliation(s)
- Patrick A Boland
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - N P Hardy
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - A Moynihan
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - P D McEntee
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - C Loo
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
| | - H Fenlon
- Department of Radiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - R A Cahill
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland.
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland.
| |
Collapse
|
9
|
Luo Q, Teng X, Dai M, Yang J, Cheng W, Chen K, Zhou L. Global trends in the application of fluorescence imaging in pancreatic diseases: a bibliometric and knowledge graph analysis. Front Oncol 2024; 14:1383798. [PMID: 39099697 PMCID: PMC11294181 DOI: 10.3389/fonc.2024.1383798] [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: 02/10/2024] [Accepted: 07/01/2024] [Indexed: 08/06/2024] Open
Abstract
Background In recent years, with the continuous development of fluorescence imaging technology, research on its application in pancreatic diseases has surged. This area is currently of high research interest and holds the potential to become a non-invasive and effective tool in the diagnosis and treatment of pancreatic diseases. The objective of this study is to explore the hotspots and trends in the field of fluorescence imaging technology applications in pancreatic diseases from 2003 to 2023 through bibliometric and visual analysis. Methods This study utilized the Web of Science (core collection) to identify publications related to the application of fluorescence imaging technology in pancreatic diseases from 2003 to 2023. Tools such as CiteSpace (V 6.2.R6), VOSviewer (v1.6.20), and R Studio (Bibliometrix: R-tool version 4.1.4) were employed to analyze various dimensions including publication count, countries, institutions, journals, authors, co-cited references, keywords, burst words, and references. Results A comprehensive analysis was conducted on 913 papers published from January 1, 2003, to December 1, 2023, on the application of fluorescence imaging technology in pancreatic diseases. The number of publications in this field has rapidly increased, with the United States being the central hub. The University of California, San Diego emerged as the most active institution. "Biomaterials" was identified as the most influential journal. Authors with the most publications and the highest average citations per article are Hoffman, Robert M. and Luiken, George A., respectively. Keywords such as pancreatic cancer, cancer, expression, indocyanine green, and nanoparticles received widespread attention, with indocyanine green and nanoparticles being current active research hotspots in the field. Conclusion This study is the first bibliometric analysis in the field of fluorescence imaging technology applications in pancreatic diseases. Our data will facilitate a better understanding of the developmental trends, identification of research hotspots, and direction in this field. The findings provide practical information for other scholars to grasp key directions and cutting-edge insights.
Collapse
Affiliation(s)
- Quanneng Luo
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Xiong Teng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - ManXiong Dai
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Jun Yang
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Hunan Schistosomiasis Control Center (Hunan Third People’s Hospital), Yueyang, Hunan, China
| | - Kang Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Lei Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| |
Collapse
|
10
|
Morales-Conde S, Navarro-Morales L, Moreno-Suero F, Balla A, Licardie E. Fluorescence and tracers in surgery: the coming future. Cir Esp 2024; 102 Suppl 1:S45-S60. [PMID: 38851317 DOI: 10.1016/j.cireng.2024.05.011] [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: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024]
Abstract
The revolution that we are seeing in the world of surgery will determine the way we understand surgical approaches in coming years. Since the implementation of minimally invasive surgery, innovations have constantly been developed to allow the laparoscopic approach to go further and be applied to more and more procedures. In recent years, we have been in the middle of another revolutionary era, with robotic surgery, the application of artificial intelligence and image-guided surgery. The latter includes 3D reconstructions for surgical planning, virtual reality, holograms or tracer-guided surgery, where ICG-guided fluorescence has provided a different perspective on surgery. ICG has been used to identify anatomical structures, assess tissue perfusion, and identify tumors or tumor lymphatic drainage. But the most important thing is that this technology has come hand in hand with the potential to develop other types of tracers that will facilitate the identification of tumor cells and ureters, as well as different light beams to identify anatomical structures. These will lead to other types of systems to assess tissue perfusion without the use of tracers, such as hyperspectral imaging. Combined with the upcoming introduction of ICG quantification, these developments represent a real revolution in the surgical world. With the imminent implementation of these technological advances, a review of their clinical application in general surgery is timely, and this review serves that aim.
Collapse
Affiliation(s)
- Salvador Morales-Conde
- Servicio de Cirugía General y Digestiva, Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain; Servicio de Cirugía General y Digestiva, Hospital Quironsalud Sagrado Corazón, Sevilla, Spain.
| | - Laura Navarro-Morales
- Servicio de Cirugía General y Digestiva, Hospital Quironsalud Sagrado Corazón, Sevilla, Spain.
| | - Francisco Moreno-Suero
- Servicio de Cirugía General y Digestiva, Hospital Quironsalud Sagrado Corazón, Sevilla, Spain.
| | - Andrea Balla
- Servicio de Cirugía General y Digestiva, Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain; Servicio de Cirugía General y Digestiva, Hospital Quironsalud Sagrado Corazón, Sevilla, Spain.
| | - Eugenio Licardie
- Servicio de Cirugía General y Digestiva, Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain; Servicio de Cirugía General y Digestiva, Hospital Quironsalud Sagrado Corazón, Sevilla, Spain.
| |
Collapse
|
11
|
Fransvea P, Chiarello MM, Fico V, Cariati M, Brisinda G. Indocyanine green: The guide to safer and more effective surgery. World J Gastrointest Surg 2024; 16:641-649. [PMID: 38577071 PMCID: PMC10989327 DOI: 10.4240/wjgs.v16.i3.641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/15/2024] [Accepted: 02/08/2024] [Indexed: 03/22/2024] Open
Abstract
In this editorial we comment on the article by Kalayarasan and co-workers published in the recent issue of the World Journal of Gastrointestinal Surgery. The authors present an interesting review on the use of indocyanine green fluorescence in different aspects of abdominal surgery. They also highlight future perspectives of the use of indocyanine green in mini-invasive surgery. Indocyanine green, used for fluorescence imaging, has been approved by the Food and Drug Administration and is safe for use in humans. It can be administered intravenously or intra-arterially. Since its advent, there have been several advancements in the applications of indocyanine green, especially in the surgical field, such as intraoperative mapping and biopsy of sentinel lymph node, measurement of hepatic function prior to resection, in neurosurgical cases to detect vascular anomalies, in cardiovascular cases for patency and assessment of vascular abnormalities, in predicting healing following amputations, in helping visualization of hepatobiliary anatomy and blood vessels, in reconstructive surgery, to assess flap viability and for the evaluation of tissue perfusion following major trauma and burns. For these reasons, the intraoperative use of indocyanine green has become common in a variety of surgical specialties and transplant surgery. Colorectal surgery has just lately begun to adopt this technique, particularly for perfusion visualization to prevent anastomotic leakage. The regular use of indocyanine green coupled with fluorescence angiography has recently been proposed as a feasible tool to help improve patient outcomes. Using the best available data, it has been shown that routine use of indocyanine green in colorectal surgery reduces the rates of anastomotic leak. The use of indocyanine green is proven to be safe, feasible, and effective in both elective and emergency scenarios. However, additional robust evidence from larger-scale, high-quality studies is essential before incorporating indocyanine green guided surgery into standard practice.
Collapse
Affiliation(s)
- Pietro Fransvea
- Emergency Surgery and Trauma Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | | | - Valeria Fico
- Emergency Surgery and Trauma Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Maria Cariati
- Department of Surgery, Azienda Sanitaria Provinciale di Crotone, Crotone 88900, Italy
| | - Giuseppe Brisinda
- Department of Abdominal and Endocrine Metabolic Medical and Surgical Sciences, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Emergency Surgery and Trauma Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| |
Collapse
|
12
|
Dai M, Qi S, Zhao X, Zhou L, Luo Q, Teng X, Cheng W, Zhou N, Liu H, Chen K. JS-K Combined with a Melanin-Based Theranostic Agent: A Novel Sequential Delivery Strategy to Enhance the Near-Infrared Fluorescence Imaging of Pancreatic Ductal Adenocarcinoma. Anal Chem 2024; 96:4103-4110. [PMID: 38427614 DOI: 10.1021/acs.analchem.3c04914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with a 5 year survival rate less than 12%. This malignancy is closely related to the unique tumor microenvironment (TME), which is characterized by a hypovascular and hyperdense extracellular matrix, making it difficult for drugs to permeate the tumor center. Near-infrared fluorescence (NIRF) imaging, which has high sensitivity and resolution, may improve the survival rate of PDAC patients. In this study, we first used JS-K (O2-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazine-1-yl] diazene-1-ium-1,2-diolate) to specifically dilate blood vessels within the TME of PDAC patients and subsequently injected IR820-PEG-MNPs (IPM NPs) to diagnose and treat orthotopic PDAC. We found that JS-K promoted the accumulation of IPM NPs in orthotopic Pan02 tumor-bearing mice and was able to increase the tumor signal-to-background ratio (SBR) in the orthotopic PDAC area by 41.5%. In addition, surgical navigation in orthotopic Pan02 tumor-bearing mice and complete tumor resection based on fluorescence imaging were achieved with a detection sensitivity of 81.0%. Moreover, we verified the feasibility of the combination of laparoscopy and photothermal ablation (PTA) for the treatment of PDAC. Finally, we demonstrated that IPM NPs had greater affinity for human PDAC tissues than for normal pancreatic tissues ex vivo, preliminarily highlighting the potential for clinical translation of these NPs. In conclusion, we developed and validated a novel sequential delivery strategy that promotes the accumulation of nanoagents in the tumor area and can be used for the diagnosis and treatment of PDAC.
Collapse
Affiliation(s)
- Manxiong Dai
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| | - Shuo Qi
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| | - Xingyang Zhao
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province 510280, P.R. China
| | - Lei Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| | - Quanneng Luo
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| | - Xiong Teng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| | - Ning Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| | - Hongwen Liu
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| | - Kang Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province 410005, P.R. China
| |
Collapse
|
13
|
Gow KW, Lautz TB, Malek MM, Cost NG, Newman EA, Dasgupta R, Christison-Lagay ER, Tiao GM, Davidoff AM. Children's Oncology Group's 2023 blueprint for research: Surgery. Pediatr Blood Cancer 2024; 71:e30766. [PMID: 37950538 PMCID: PMC10872730 DOI: 10.1002/pbc.30766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Surgery plays a crucial role in the treatment of children with solid malignancies. A well-conducted operation is often essential for cure. Collaboration with the primary care team is important for determining if and when surgery should be performed, and if performed, an operation must be done in accordance with well-established standards. The long-term consequences of surgery also need to be considered. Indications and objectives for a procedure vary. Providing education and developing and analyzing new research protocols that include aims relevant to surgery are key objectives of the Surgery Discipline of the Children's Oncology Group. The critical evaluation of emerging technologies to ensure safe, effective procedures is another key objective. Through research, education, and advancing technologies, the role of the pediatric surgeon in the multidisciplinary care of children with solid malignancies will continue to evolve.
Collapse
Affiliation(s)
- Kenneth W. Gow
- Division of General & Thoracic Surgery, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Timothy B. Lautz
- Department of Pediatric Surgery, Lurie Children’s Hospital of Chicago, Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Marcus M. Malek
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nicholas G. Cost
- Department of Surgery, Division of Urology and the Surgical Oncology Program, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Erika A. Newman
- Department of Surgery, Mott Children’s Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Roshni Dasgupta
- Division of Pediatric General and Thoracic Surgery, Cincinnati Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Emily R. Christison-Lagay
- Division of Pediatric Surgery, Yale School of Medicine, Yale-New Haven Children’s Hospital, New Haven, Connecticut, USA
| | - Gregory M. Tiao
- Division of Pediatric General and Thoracic Surgery, Cincinnati Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Andrew M. Davidoff
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
14
|
Huang H, He S, Wei R, Zhu X, Deng Z, Wang Y, Guo L, Lei J, Cai L, Xie Y. Near-infrared (NIR) imaging with indocyanine green (ICG) may assist in intraoperative decision making and improving surgical margin in bone and soft tissue tumor surgery. J Surg Oncol 2023; 128:612-627. [PMID: 37178368 DOI: 10.1002/jso.27306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND AND OBJECTIVES Negative surgical margins are significant in improving patient outcomes. However, surgeons can only rely on visual and tactile information to identify tumor margins intraoperatively. We hypothesized that intraoperative fluorescence imaging with indocyanine green (ICG) could serve as an assistive technology to evaluate surgical margins and guide surgery in bone and soft tissue tumor surgery. METHODS Seventy patients with bone and soft tissue tumors were enrolled in this prospective, non-randomized, single-arm feasibility study. All patients received intravenous indocyanine green (0.5 mg/kg) before surgery. Near-infrared (NIR) imaging was performed on in situ tumors, wounds, and ex vivo specimens. RESULTS 60/70 tumors were fluorescent at NIR imaging. The final surgical margins were positive in 2/55 cases, including 1/40 of the sarcomas. Surgical decisions were changed in 19 cases by NIR imaging, and in 7/19 cases final pathology demonstrated margins were improved. Fluorescence analysis showed that the tumor-to-background ratio (TBR) of primary malignant tumors was higher than that of benign, borderline, metastatic, and tumors ≥5 cm in size had higher TBR than those <5 cm. CONCLUSIONS ICG fluorescence imaging may be a beneficial technique to assist in surgical decision making and improving surgical margins in bone and soft tissue tumor surgery.
Collapse
Affiliation(s)
- Huayi Huang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Siyuan He
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Renxiong Wei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Xiaobin Zhu
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Zhouming Deng
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yi Wang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Liangyu Guo
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Jun Lei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yuanlong Xie
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
15
|
Husarova T, MacCuaig WM, Dennahy IS, Sanderson EJ, Edil BH, Jain A, Bonds MM, McNally MW, Menclova K, Pudil J, Zaruba P, Pohnan R, Henson CE, Grizzle WE, McNally LR. Intraoperative Imaging in Hepatopancreatobiliary Surgery. Cancers (Basel) 2023; 15:3694. [PMID: 37509355 PMCID: PMC10377919 DOI: 10.3390/cancers15143694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Hepatopancreatobiliary surgery belongs to one of the most complex fields of general surgery. An intricate and vital anatomy is accompanied by difficult distinctions of tumors from fibrosis and inflammation; the identification of precise tumor margins; or small, even disappearing, lesions on currently available imaging. The routine implementation of ultrasound use shifted the possibilities in the operating room, yet more precision is necessary to achieve negative resection margins. Modalities utilizing fluorescent-compatible dyes have proven their role in hepatopancreatobiliary surgery, although this is not yet a routine practice, as there are many limitations. Modalities, such as photoacoustic imaging or 3D holograms, are emerging but are mostly limited to preclinical settings. There is a need to identify and develop an ideal contrast agent capable of differentiating between malignant and benign tissue and to report on the prognostic benefits of implemented intraoperative imaging in order to navigate clinical translation. This review focuses on existing and developing imaging modalities for intraoperative use, tailored to the needs of hepatopancreatobiliary cancers. We will also cover the application of these imaging techniques to theranostics to achieve combined diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Tereza Husarova
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - William M. MacCuaig
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Isabel S. Dennahy
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Emma J. Sanderson
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Barish H. Edil
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Ajay Jain
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Morgan M. Bonds
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Molly W. McNally
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Katerina Menclova
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - Jiri Pudil
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - Pavel Zaruba
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - Radek Pohnan
- Department of Surgery, Military University Hospital Prague, 16902 Prague, Czech Republic
| | - Christina E. Henson
- Department of Radiation Oncology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lacey R. McNally
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
16
|
Harris AC, Choudhury S, Pachl M. Early results of minimally invasive fluorescent guided pediatric oncology surgery with delivery of indocyanine green during induction of anesthesia. Photodiagnosis Photodyn Ther 2023; 42:103639. [PMID: 37245684 DOI: 10.1016/j.pdpdt.2023.103639] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND Indocyanine green (ICG) fluoresces in the near infra-red (NIR) spectrum. It is widely used in adult oncological surgery for identification of tumor margins and lymph node sampling. However, deliver of ICG in almost all studies is 24 h or more prior to surgery. This is the first study in children to assess its feasibility in minimally invasive surgery (MIS) for oncological disease following ICG injection during induction of anesthesia. METHODS This was an open label, prospective, single center, feasibility study recruiting consecutive patients eligible for MIS tumor resection or metastectomy. ICG was injected intravenously at induction of anesthesia. Patient demographics, intraoperative appearances, post-operative histopathology, and surgeon Likert ratings were collected. RESULTS Fourteen patients were included. Five had lung metastases (Wilms, Osteosarcoma (2), Hodgkin's, melanoma) and 9 had other tumors (neuroblastoma, inflammatory myofibroblastic tumor, ganglioneuroma, phaeochromocytoma, adrenal tumor). Lung metastases were easily identifiable, and all had negative margins. Tumors containing viable disease fluoresced and were completely resected, whilst benign and heavily treated tumors were afluorescent. There were no adverse events relating to ICG or issues with background fluorescence. CONCLUSION Based on this small sample, injection of ICG during induction of anesthesia is safe and effective in showing tumor margins in patients who have had little or no neoadjuvant chemotherapy as well as in metastectomy in Wilms and osteosarcoma. Further studies are needed to confirm these preliminary results.
Collapse
Affiliation(s)
- Anna Cv Harris
- Department of Paediatric Surgery and Urology, Birmingham Women's, and Children's NHS Foundation Trust, UK
| | - S Choudhury
- Department of Paediatric Surgery and Urology, Birmingham Women's, and Children's NHS Foundation Trust, UK
| | - Max Pachl
- Department of Paediatric Surgery and Urology, Birmingham Women's, and Children's NHS Foundation Trust, UK; Cancer Research UK Clinical Trials Unit, Institute of Cancer and Genomic Sciences, University of Birmingham, UK.
| |
Collapse
|
17
|
Liu J, Xu Z, Meng C, Wusiman S, Xie X, Wang Y, Xiao F, Gu C, Chen J, Ling CC, Li P, Yuan Z, Ling Y. Acidic tumor microenvironment-activatable fluorescent diagnostic probe for the rapid identification and resection of human tumors via spraying. Biosens Bioelectron 2023; 234:115343. [PMID: 37167656 DOI: 10.1016/j.bios.2023.115343] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/16/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023]
Abstract
A fluorescent diagnostic probe for real-time intraoperative image-guided tumor resection can significantly improve the efficiency and quality of oncological therapy, but their development is challenging. Herein, a novel fluorescent diagnostic probe called HLTC based on β-carboline was designed and synthesized. HLTC was found to show a ∼10-fold enhancement of fluorescence quantum field with pH from 7.4 to 4.0, indicating its imaging potential in acid environment which is a typical hallmark of the tumor microenvironment (TME). Following fluorescence microscopy imaging showed HLTC could emit specific signals in cancer cells and sections, by both one-photon excitation and two-photon excitation. Importantly, HLTC enabled the precise and rapid delineation of both transplanted tumor and clinical tumor tissues within several minutes of simple topical spray. The tumor-to-background ratio (TBR) was up to 10.2 ± 1.0 at clinical liver cancer tissues and 9.9 ± 0.3 at clinical colon cancer tissues, allowing precise tumor margin identification and the effective guidance of surgical tumor resection. Furthermore, CCK8 assay, pharmacokinetic evaluation, blood analysis and H&E staining were performed, which verified high biocompatibility and biosafety of HLTC at working concentration. These results reveal the exciting potential of this small-molecule fluorescent diagnostic probe for real-time fluorescence-based navigation during surgical tumor resection.
Collapse
Affiliation(s)
- Ji Liu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China; Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing, 210009, China
| | - Zhongyuan Xu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Chi Meng
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Sainaiwaiergul Wusiman
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing, 210009, China
| | - Xudong Xie
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Yichen Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China
| | - Feng Xiao
- Department of Pathology, Nantong Third People's Hospital and the Third Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Chunyan Gu
- Department of Pathology, Nantong Third People's Hospital and the Third Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jun Chen
- Department of Hepatobiliary Surgery, Nantong Third People's Hospital and the Third Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Chang-Chun Ling
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Peng Li
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, 639 Longmian Road, Jiangning District, Nanjing, 210009, China.
| | - Yong Ling
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, 226001, China.
| |
Collapse
|
18
|
Lu WL, Kuang H, Gu J, Hu X, Chen B, Fan Y. GAP-43 targeted indocyanine green-loaded near-infrared fluorescent probe for real-time mapping of perineural invasion lesions in pancreatic cancer in vivo. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102671. [PMID: 37054805 DOI: 10.1016/j.nano.2023.102671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/22/2023] [Accepted: 03/26/2023] [Indexed: 04/15/2023]
Abstract
OBJECTIVE Perineural invasion (PNI) is associated with local recurrence, distant metastasis, and a poor prognosis in pancreatic cancer. However, rare attempt was made to identified the PNI intraoperative. To facilitate precise R0 excision of the tumor, we planned to develop a fluorescent probe for intraoperative imaging of the PNI using GAP-43 as the target and indocyanine green (ICG) as the carrier. METHODS The probe was created by binding peptide antibody and ICG. Its targeting was tested in vitro and in vivo using a co-culture model of PC12 and tumor cells to create an in vitro neural invasion model and a mouse sciatic nerve invasion model. The small animal imaging system and surgical navigation system confirmed the probe's potential clinical applicability. The sciatic nerve damage model was created to confirm the probe's targeting. RESULTS We used the pancreatic cancer samples and the public database to confirm that GAP-43 was preferentially overexpressed in pancreatic cancer, particularly in PNI. PC12 cells showed high GAP-43RA-PEG-ICG probe-specific absorption after being co-cultured with tumor cells in vitro. In the sciatic nerve invasion experiment, animals in probe group displayed a significantly stronger fluorescence signal at the PNI compared to ICG-NP and the contralateral normal nerves groups. Although only 60 % of mice appeared to have R0 resections by the naked eye, small animal imaging systems and surgical fluorescence navigation systems could remove the tumor with R0 precision. The injury model used in the probe imaging experimental trials demonstrated that the probe was specifically targeted to the injured nerve, regardless of whether the injury was infiltrated by a tumor or physical. CONCLUSION We developed the GAP-43Ra-ICG-PEG, an active-targeting near-infrared fluorescent (NIF) probe, that specifically binds to GAP-43-positive neural cells in an in vitro model of PNI. The probe efficiently visualized PNI lesions in pancreatic cancer in preclinical models, opening up new possibilities for NIRF-guided pancreatic surgery, particularly for PNI patients.
Collapse
Affiliation(s)
- Wen Liang Lu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Thyroid and breast surgery, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China
| | - Houfang Kuang
- Department of General Surgery, Wuhan Children(,) hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China
| | - Jianyou Gu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaojun Hu
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Department of Hepatobiliary Surgery, The Fifth Affifiliated Hospital of Southern Medical University, Guangzhou 510920, China
| | - Bo Chen
- Department of Thyroid and breast surgery, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China
| | - Yingfang Fan
- The Department of General Surgery & Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
19
|
Tao H, Zeng X, Lin W, Wang Z, Lin J, Li J, Qian Y, Yang J, Fang C. Indocyanine green fluorescence imaging to localize insulinoma and provide three-dimensional demarcation for laparoscopic enucleation: a retrospective single-arm cohort study. Int J Surg 2023; 109:821-828. [PMID: 37026828 PMCID: PMC10389620 DOI: 10.1097/js9.0000000000000319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/20/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Indocyanine green (ICG) fluorescence imaging is a new technology that can improve the real-time location of tumor edges and small nodules during surgery. However, no study has investigated its application in laparoscopic insulinoma enucleation. This study aimed to evaluate the feasibility and accuracy of this method for intraoperative localization of insulinomas and margin assessment during laparoscopic insulinoma enucleation. MATERIALS AND METHODS Eight patients who underwent laparoscopic insulinoma enucleation from October 2016 to June 2022 were enrolled. Two methods of ICG administration, ICG dynamic perfusion and three-dimensional (3D) demarcation staining, were utilized in the laparoscopic insulinoma enucleation. Tumor-to-background ratio (TBR) and histopathologic analysis were used to evaluate the feasibility and accuracy of these novel navigation methods in laparoscopic insulinoma enucleation. RESULTS All eight enrolled patients underwent both ICG dynamic perfusion and 3D demarcation staining. ICG dynamic perfusion images were available for six of them, among which five tumors could be recognized by TBR (largest TBR in each case 4.42±2.76), while the other could be distinguished by the disordered blood vessels in the tumor area. Seven out of eight specimens had successful 3D demarcation staining (TBR 7.62±2.62). All wound bed margins had negative frozen sections and final histopathologic diagnoses. CONCLUSIONS ICG dynamic perfusion may be helpful in observing the abnormal vascular perfusion of tumors, providing similar functionality to intraoperative real-time angiography. ICG injection under the tumor pseudocapsule may be a useful method for acquiring real-time, 3D demarcation for the resection of insulinoma.
Collapse
Affiliation(s)
- Haisu Tao
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou
| | - Xiaojun Zeng
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou
| | - Wenjun Lin
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou
| | - Zhuangxiong Wang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou
| | - Jinyu Lin
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou
| | - Jiang Li
- The First Affiliated Hospital, College of Medicine, Shihezi University, Shihezi
| | - Yinling Qian
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen
| | - Jian Yang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou
- Pazhou Lab, Guangzhou, People’s Republic of China
| | - Chihua Fang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University
- Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou
- Pazhou Lab, Guangzhou, People’s Republic of China
| |
Collapse
|
20
|
Cassinotti E, Al-Taher M, Antoniou SA, Arezzo A, Baldari L, Boni L, Bonino MA, Bouvy ND, Brodie R, Carus T, Chand M, Diana M, Eussen MMM, Francis N, Guida A, Gontero P, Haney CM, Jansen M, Mintz Y, Morales-Conde S, Muller-Stich BP, Nakajima K, Nickel F, Oderda M, Parise P, Rosati R, Schijven MP, Silecchia G, Soares AS, Urakawa S, Vettoretto N. European Association for Endoscopic Surgery (EAES) consensus on Indocyanine Green (ICG) fluorescence-guided surgery. Surg Endosc 2023; 37:1629-1648. [PMID: 36781468 PMCID: PMC10017637 DOI: 10.1007/s00464-023-09928-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 01/28/2023] [Indexed: 02/15/2023]
Abstract
BACKGROUND In recent years, the use of Indocyanine Green (ICG) fluorescence-guided surgery during open and laparoscopic procedures has exponentially expanded across various clinical settings. The European Association of Endoscopic Surgery (EAES) initiated a consensus development conference on this topic with the aim of creating evidence-based statements and recommendations for the surgical community. METHODS An expert panel of surgeons has been selected and invited to participate to this project. Systematic reviews of the PubMed, Embase and Cochrane libraries were performed to identify evidence on potential benefits of ICG fluorescence-guided surgery on clinical practice and patient outcomes. Statements and recommendations were prepared and unanimously agreed by the panel; they were then submitted to all EAES members through a two-rounds online survey and results presented at the EAES annual congress, Barcelona, November 2021. RESULTS A total of 18,273 abstracts were screened with 117 articles included. 22 statements and 16 recommendations were generated and approved. In some areas, such as the use of ICG fluorescence-guided surgery during laparoscopic cholecystectomy, the perfusion assessment in colorectal surgery and the search for the sentinel lymph nodes in gynaecological malignancies, the large number of evidences in literature has allowed us to strongly recommend the use of ICG for a better anatomical definition and a reduction in post-operative complications. CONCLUSIONS Overall, from the systematic literature review performed by the experts panel and the survey extended to all EAES members, ICG fluorescence-guided surgery could be considered a safe and effective technology. Future robust clinical research is required to specifically validate multiple organ-specific applications and the potential benefits of this technique on clinical outcomes.
Collapse
Affiliation(s)
- E Cassinotti
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, University of Milan, Via Francesco Sforza 35, 20121, Milan, Italy.
| | - M Al-Taher
- Research Institute Against Digestive Cancer (IRCAD), Strasbourg, France
| | - S A Antoniou
- Department of Surgery, Papageorgiou General Hospital, Thessaloniki, Greece
| | - A Arezzo
- Department of Surgical Sciences, University of Torino, Turin, Italy
| | - L Baldari
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, University of Milan, Via Francesco Sforza 35, 20121, Milan, Italy
| | - L Boni
- Department of General and Minimally Invasive Surgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico di Milano, University of Milan, Via Francesco Sforza 35, 20121, Milan, Italy
| | - M A Bonino
- Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - N D Bouvy
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - R Brodie
- Department of General Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - T Carus
- Niels-Stensen-Kliniken, Elisabeth-Hospital, Thuine, Germany
| | - M Chand
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences (WEISS), University College London, London, UK
- Division of Surgery and Interventional Sciences, University College London, London, UK
| | - M Diana
- IHU Strasbourg, Institute of Image-Guided Surgery and IRCAD, Research Institute Against Cancer of the Digestive System, Strasbourg, France
| | - M M M Eussen
- Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - N Francis
- Department of General Surgery, Yeovil District Hospital NHS Foundation Trust, Yeovil, UK
| | - A Guida
- Department of Medico-Surgical Sciences and Translation Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - P Gontero
- Division of Urology, Department of Surgical Science, AOU Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - C M Haney
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - M Jansen
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Y Mintz
- Department of General Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - S Morales-Conde
- Unit of Innovation in Minimally Invasive Surgery, Department of General Surgery, University Hospital Virgen del Rocío, University of Sevilla, Seville, Spain
| | - B P Muller-Stich
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - K Nakajima
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - F Nickel
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - M Oderda
- Division of Urology, Department of Surgical Science, AOU Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - P Parise
- U.O.C. Chirurgia Generale, Policlinico di Abano Terme, Abano Terme, PD, Italy
| | - R Rosati
- Department of Gastrointestinal Surgery, San Raffaele Hospital, Milan, Italy
| | - M P Schijven
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, North Holland, The Netherlands
- Amsterdam Gastroenterology and Metabolism, Amsterdam UMC, Amsterdam, North Holland, The Netherlands
- Amsterdam Public Health, Digital Health, Amsterdam UMC, Amsterdam, North Holland, The Netherlands
| | - G Silecchia
- Department of Medico-Surgical Sciences and Translation Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - A S Soares
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences (WEISS), University College London, London, UK
- Division of Surgery and Interventional Sciences, University College London, London, UK
| | - S Urakawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - N Vettoretto
- U.O.C. Chirurgia Generale, ASST Spedali Civili di Brescia P.O. Montichiari, Ospedale di Montichiari, Montichiari, Italy
| |
Collapse
|
21
|
Li P, Liu J, He K, Gong S, Chi C, Liu P, Su G, Li W, Duan H, Liu P, Tian J, Chen C. Tumor lesion detection in patients with cervical cancer by indocyanine green near-infrared imaging. Eur J Nucl Med Mol Imaging 2023; 50:1252-1261. [PMID: 36450938 DOI: 10.1007/s00259-022-06030-1] [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: 08/08/2022] [Accepted: 10/30/2022] [Indexed: 12/03/2022]
Abstract
PURPOSE To investigate the feasibility and accuracy of near-infrared fluorescence (NIRF) imaging for detecting the extent of tumor invasion in cervical cancer using indocyanine green (ICG). METHODS We enrolled 51 patients who were diagnosed with cervical cancer with FIGO stage IB1-IIA2 disease. Patients were administered indocyanine green (ICG) at a dose of 5 mg/kg 24 h prior to surgery. A customized near-infrared fluorescence (NIRF) imaging system was used to identify the extent of tumor invasion when radical hysterectomy specimens were harvested. The relationship between tumor fluorescence intensity and clinicopathological characteristics was analyzed. RESULTS Of the 51 enrolled patients, 3 patients did not have residual tumors after cervical conization, and tumor lesions were identified by NIRF imaging in all the remaining 48 patients. The results of NIRF imaging were in agreement with the postoperative pathological findings in 95.8% of the patients with stromal invasion, 100% of those with surgical margin invasion, 100% of those with parametrial tumor involvement, and 100% of patients with uterine corpus invasion. The mean signal-to-background ratio (SBR) of the cervical tumors was 2.91 ± 1.64, and the SBR was independent of clinicopathological characteristics. Fluorescence microscopy confirmed that ICG fluorescence was present in the tumor nests. CONCLUSIONS NIRF imaging enables objective, accurate, and safe identification of tumor invasion during cervical cancer surgery. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov NCT04224467.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue, Guangzhou, 510515, China
| | - Jiaqi Liu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue, Guangzhou, 510515, China
| | - Kunshan He
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, 100191, China.,Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, 100191, Beijing, 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, 100190, China
| | - Shipeng Gong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue, Guangzhou, 510515, China
| | - Chongwei Chi
- 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, 100190, China
| | - Pan Liu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue, Guangzhou, 510515, China
| | - Guidong Su
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, 100191, China
| | - Weili Li
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue, Guangzhou, 510515, China
| | - Hui Duan
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue, Guangzhou, 510515, China
| | - Ping Liu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue, Guangzhou, 510515, China.
| | - Jie Tian
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing, 100191, China. .,Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, 100191, Beijing, 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, 100190, China.
| | - Chunlin Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue, Guangzhou, 510515, China.
| |
Collapse
|
22
|
Zhu J, Jiang Y, Pan X, Xu K, Niu W, Lv Y, Li C, Wang Y, Xue Z, Lei P, He Y. In Vivo Evaluation of a Gallium-68-Labeled Tumor-Tracking Cyanine Dye for Positron Emission Tomography/Near-Infrared Fluorescence Carcinoma Imaging, Image-Guided Surgery, and Photothermal Therapy. ACS OMEGA 2023; 8:6067-6077. [PMID: 36816684 PMCID: PMC9933465 DOI: 10.1021/acsomega.2c08235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
Positron emission tomography (PET)/near-infrared fluorescence (NIRF) dual-modal imaging presents an enticing prospect for tumor diagnosis and surgical navigation. In this study, we developed a novel probe IR808-DOTA for tumor-targeted PET/NIRF imaging, image-guided surgery, and photothermal therapy. This construct had better water solubility and pharmacokinetics than IR808 and had similar photophysical properties, tumor targeting ability, and photothermal anticancer effect to IR808. By a simple labeling process, IR808-DOTA was labeled with gallium-68 and applied as a PET probe for tumor imaging in MCF-7 tumor xenografted mice. IR808-DOTA itself acted as an NIRF imaging agent in the following surgery for intraoperative navigation to aid surgeons in the delineation of tumor margins and visualizing sentinel lymph nodes to facilitate a more thorough tumor resection. Irradiation by laser, IR808-DOTA could prominently inhibit tumor growth in MCF-7 subcutaneous tumor model mice by directly ablating tumor cells, inhibiting tumor proliferation, and promoting tumor cell apoptosis. In summary, 68Ga-DOTA-IR808 could enable a convenient and user-friendly workflow for tumor imaging and guided surgery, and therefore, it may have great prospects for clinical translation as a PET/NIRF dual-modal probe.
Collapse
Affiliation(s)
- Jiaxu Zhu
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yaqun Jiang
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xin Pan
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Kui Xu
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Wenhao Niu
- Department
of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yibing Lv
- Department
of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Chongjiao Li
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yichun Wang
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Zejian Xue
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| | - Ping Lei
- Department
of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Yong He
- Department
of Nuclear Medicine, Zhongnan Hospital of
Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
23
|
Azari F, Kennedy G, Bernstein E, Delikatny J, Lee JYK, Kucharczuk J, Low PS, Singhal S. Evaluation of OTL38-Generated Tumor-to-Background Ratio in Intraoperative Molecular Imaging-Guided Lung Cancer Resections. Mol Imaging Biol 2023; 25:85-96. [PMID: 34101106 PMCID: PMC8651846 DOI: 10.1007/s11307-021-01618-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Cancer surgery has multiple challenges including localizing small lesions, ensuring negative margins, and identifying synchronous cancers. One of the tools proposed to address these issues is intraoperative molecular imaging (IMI). An important consideration in IMI is the quantification of the tumor fluorescence during the procedure and using that data to add clinical value. Currently, the most commonly cited measure of quantification is the tumor-to-background ratio (TBR). Our goal was to evaluate the clinical value of TBR measured with OTL38 NIR tracer during a lung cancer resection. METHODS Intraoperative data was retrospectively reviewed from a prospectively collected 5-year database. Between 2015 and 2020, 279 patients were included in the study. For standardization, all patients underwent infusion of the same targeted molecular optical contrast agent (OTL38) for lung cancer resections; then, the mean fluorescence intensity of the tumors and background tissues were calculated. To evaluate the clinical efficacy of the TBR calculation, the results were correlated with patient, biologic, tumor, and technological factors. RESULTS For pulmonary surgery, patient factors such as gender, age, smoking history, and time from infusion of OTL38 to surgery did not have any statistical significance in predicting the TBR during surgery. In addition, TBR measurements did not correlate with location of the tumor in the lung (p = 0.123). There was no statistical correlation of preoperative positron emission tomography measurements (standardized uptake value) with intraoperative TBR. However, there was statistically significant negative correlation of in situ TBR measurement and the distance of the lesion from the surface of the organ (p < 0.001). Adenocarcinoma spectrum lesions overall had statistically significant correlation with in situ fluorescence compared to other NSCLC malignancies (p < 0.01) but TBR measurements could not identify histopathologic subtype on univariate analysis (p = 0.089). There was a tendency for in situ fluorescence for moderately and well-differentiated adenocarcinoma spectrum lesions, but this was not statistically significant. When comparing the in situ TBR of benign to malignant nodules in the lung, there was no statistically significant association (p = 0.145). In subset analysis, adenocarcinoma spectrum lesions tend to fluoresce at brighter with OTL38 compared to other histologic subtypes. CONCLUSION In our various iterations, the results of our retrospective analysis did not show that TBR measurements during OTL38-guided surgery provide clinically useful information about the nature of the nodule or cancer. The true value of IMI is in the ability for the surgeon to use the fluorescence to guide the surgeon to the tumor and margins, but that sophisticated quantification of the amount of fluorescence may not have clinical utility.
Collapse
Affiliation(s)
- Feredun Azari
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA
| | - Gregory Kennedy
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA
| | - Elizabeth Bernstein
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA
| | - James Delikatny
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John Kucharczuk
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA
| | - Phil S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA.
| |
Collapse
|
24
|
Jia D, Liu H, Zheng S, Yuan D, Sun R, Wang F, Li Y, Li H, Yuan F, Fan Q, Zhao Z. ICG-Dimeric Her2-Specific Affibody Conjugates for Tumor Imaging and Photothermal Therapy for Her2-Positive Tumors. Mol Pharm 2023; 20:427-437. [PMID: 36315025 DOI: 10.1021/acs.molpharmaceut.2c00708] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Human epidermal growth factor receptor 2 (Her2) is abundantly expressed in various solid tumors. The Her2-specific Affibody (ZHer2:2891) has been clinically tested in patients with Her2-positive breast cancer and is regarded as an ideal drug carrier for tumor diagnosis and targeted treatment. Indocyanine green (ICG) can be used as a photosensitizer for photothermal therapy (PTT), in addition to fluorescent dyes for tumor imaging. In this study, a dimeric Her2-specific Affibody (ZHer2) based on ZHer2:2891 was prepared using the E. coli expression system and then coupled to ICG through an N-hydroxysuccinimide (NHS) ester reactive group to construct a novel bifunctional protein drug (named ICG-ZHer2) for tumor diagnosis and PTT. In vitro, ICG-ZHer2-mediated PTT selectively and efficiently killed Her2-positive BT-474 and SKOV-3 tumor cells rather than Her2-negative HeLa tumor cells. In vivo, ICG-ZHer2 specifically accumulated in Her2-positive SKOV-3 tumor grafts rather than Her2-negative HeLa tumor grafts; high-contrast tumor optical images were obtained. However, Her2-negative HeLa tumor grafts were not detected. More importantly, ICG-ZHer2-mediated PTT exhibited a significantly enhanced antitumor effect in mice bearing SKOV-3 tumor grafts owing to the good photothermal properties of ICG-ZHer2. Of note, ICG-ZHer2 did not exhibit acute toxicity in mice during short-term treatment. Overall, our findings indicate that ICG-ZHer2 is a promising bifunctional drug for Her2-positive tumor diagnosis and PTT.
Collapse
Affiliation(s)
- Dianlong Jia
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Huimin Liu
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Shuhui Zheng
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Dandan Yuan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Ruohan Sun
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Fei Wang
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Yang Li
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Hui Li
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zhenxiong Zhao
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 317700, China
| |
Collapse
|
25
|
Wang J, Li S, Wang K, Zhu L, Yang L, Zhu Y, Zhang Z, Hu L, Yuan Y, Fan Q, Ren J, Yang G, Ding W, Zhou X, Cui J, Zhang C, Yuan Y, Huang R, Tian J, Tao X. A c-MET-Targeted Topical Fluorescent Probe cMBP-ICG Improves Oral Squamous Cell Carcinoma Detection in Humans. Ann Surg Oncol 2023; 30:641-651. [PMID: 36184713 PMCID: PMC9726820 DOI: 10.1245/s10434-022-12532-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The postoperative survival of oral squamous cell carcinoma (SCC) relies on precise detection and complete resection of original tumors. The mucosal extension of the tumor is evaluated visually during surgery, but small and flat foci are difficult to detect. Real-time fluorescence imaging may improve detection of tumor margins. MATERIALS AND METHODS In the current study, a peptide-based near-infrared (NIR) fluorescence dye, c-MET-binding peptide-indocyanine green (cMBP-ICG), which specifically targets tumor via c-MET binding, was synthetized. A prospective pilot clinical trial then was conducted with oral SCC patients and intraoperatively to assess the feasibility of cMBP-ICG used to detect tumors margins. Fluorescence was histologically correlated to determine sensitivity and specificity. RESULTS The immunohistochemistry (IHC) results demonstrated increased c-Met expression in oral SCC compared with normal mucosa. Tumor-to-background ratios ranged from 2.71 ± 0.7 to 3.11 ± 1.2 in different concentration groups. From 10 patients with oral SCC, 60 specimens were collected from tumor margins. The sensitivity and specificity of discriminative value derived from cMBP-ICG application in humans were respectively 100% and 75%. CONCLUSIONS Topical application of cMBP-ICG is feasible and safe for optimizing intraoperative visualization and tumor margin detection in oral SCC patients, which could clinically increase the probability of complete resections and improve oncologic outcomes.
Collapse
Affiliation(s)
- Jingbo Wang
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siyi Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Ling Zhu
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Yang
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yunjing Zhu
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhen Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Longwei Hu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Yuan
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Fan
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiliang Ren
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gongxin Yang
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weilong Ding
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyu Zhou
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Junqi Cui
- Department of Pathology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunye Zhang
- Department of Pathology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Yuan
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruimin Huang
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Xiaofeng Tao
- Department of Radiology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
26
|
Wang Y, Jiao W, Yin Z, Zhao W, Zhao K, Zhou Y, Fang R, Dong B, Chen B, Wang Z. Application of near-infrared fluorescence imaging in the accurate assessment of surgical margins during breast-conserving surgery. World J Surg Oncol 2022; 20:357. [PMID: 36352391 PMCID: PMC9644613 DOI: 10.1186/s12957-022-02827-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022] Open
Abstract
Objective To evaluate the feasibility and accuracy of near-infrared fluorescence imaging technology for assessing margins during breast-conserving surgery for breast cancer. Methods Forty-three breast cancer patients who received surgical treatment at Yijishan Hospital of Wannan Medical College were selected. Before the operation, the patients were administered with an indocyanine green injection of 0.5 mg/kg intravenously 2 h before operation. During and after the operation, all patients underwent surgical margin monitoring with the near-infrared fluorescence imaging system for fluorescence imaging and acquisition of images and quantitative fluorescence intensity. During the operation, the patients’ tissue specimens were collected on the upper, lower, inner, outer, apical, and basal sides of the fluorescence boundary of the isolated lesions for pathological examination. Results Fluorescence was detected in the primary tumor in all patients. The average fluorescence intensities of tumor tissue, peritumoral tissue, and normal tissue were 219.41 ± 32.81, 143.35 ± 17.37, and 105.77 ± 17.79 arbitrary units, respectively (P < 0.05, t test). The signal-to-background ratio of tumor to peritumor tissue and normal tissue was 1.54 ± 0.20 and 2.14 ± 0.60, respectively (P < 0.05, t test). Abnormal indocyanine green fluorescence was detected in 11.6% patients (5/43), including 3 patients with residual infiltrating carcinoma and 2 patients with adenosis with ductal dilatation. Conclusion This study confirms the high sensitivity and specificity of near-infrared fluorescence imaging technology for breast-conserving surgery margin assessment. Near-infrared fluorescence imaging technology can be used as an intraoperative diagnosis and treatment tool to accurately determine the surgical margin and is of important guiding value in breast-conserving surgery for breast cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-022-02827-4.
Collapse
|
27
|
Rompianesi G, Montalti R, Giglio MC, Ceresa CDL, Nasto RA, De Simone G, Troisi RI. Systematic review, meta-analysis and single-centre experience of the diagnostic accuracy of intraoperative near-infrared indocyanine green-fluorescence in detecting pancreatic tumours. HPB (Oxford) 2022; 24:1823-1831. [PMID: 35654671 DOI: 10.1016/j.hpb.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/20/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND During pancreatic resections assessing tumour boundaries and identifying the ideal resection margins can be challenging due to the associated pancreatic gland inflammation and texture. This is particularly true in the context of minimally invasive surgery, where there is a very limited or absent tactile feedback. Indocyanine green (ICG) fluorescence imaging can assist surgeons by simply providing valuable real-time intraoperative information at low cost with minimal side effects. This meta-analysis summarises the available evidence on the use of near-infrared fluorescence imaging with ICG for the intraoperative visualization of pancreatic tumours (PROSPERO ID: CRD42021247203). METHODS MEDLINE, Embase, and Web Of Science electronic databases were searched to identify manuscripts where ICG was intravenously administered prior to or during pancreatic surgery and reporting the prevalence of pancreatic lesions visualised through fluorescence imaging. RESULTS Six studies with 7 series' reporting data on 64 pancreatic lesions were included in the analysis. MINOR scores ranged from 6 to 10, with a median of 8. The most frequent indications were pancreatic adenocarcinoma and neuroendocrine tumours. In most cases (67.2%) ICG was administered during surgery. ICG fluorescence identified 48/64 lesions (75%) with 81.3% accuracy, 0.788 (95%CI 0.361-0.961) sensitivity, 1 (95%CI 0.072-1) specificity and positive predictive value of 0.982 (95%CI 0.532-1). In line with the literature, ICG fluorescence identified 5/6 (83.3%) of pancreatic lesions during robotic pancreatic resections performed at our Institution. CONCLUSION This meta-analysis is the first summarising the results of ICG immunofluorescence in detecting pancreatic tumours during surgery, showing good accuracy. Additional research is needed to define optimal ICG administration strategies and fluorescence intensity cut-offs.
Collapse
Affiliation(s)
- Gianluca Rompianesi
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive, Robotic and Transplant Surgery, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples, Italy.
| | - Roberto Montalti
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive, Robotic and Transplant Surgery, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples, Italy
| | - Mariano C Giglio
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive, Robotic and Transplant Surgery, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples, Italy
| | - Carlo D L Ceresa
- Nuffield Department of Surgical Sciences, University of Oxford, UK
| | - Riccardo A Nasto
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive, Robotic and Transplant Surgery, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples, Italy
| | - Giuseppe De Simone
- Division of Anaesthesia of Hepato-Bilio-Pancreatic and Liver Transplant Surgery, Department of General Surgery and Surgical Specialties, Kidney Transplantation, Intensive Care and Nephrology, Federico II University Hospital, Naples, Italy
| | - Roberto I Troisi
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive, Robotic and Transplant Surgery, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples, Italy
| |
Collapse
|
28
|
Zhang Y, Zhang J, Jiang K, Wu W. Indocyanine green real-time-guided laparoscopic duodenum-preserving pancreatic head resection. J Minim Access Surg 2022; 18:632-634. [PMID: 36124475 PMCID: PMC9632711 DOI: 10.4103/jmas.jmas_205_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Duodenum-preserving pancreatic head resection (DPPHR) is very complicated due to its difficulty to find the lower common bile duct (CBD), and to preserve the blood supply of the duodenum and CBD. Recently, indocyanine green (ICG) has been widely applied for navigation during biliary system and liver surgery. However, the application of ICG-guided laparoscopic DPPHR has not been established. Herein, we report an intraoperative angiography technique using ICG fluorescence imaging to visualise blood flow, tissue perfusion, CBD navigation and bile leakage assessment.
Collapse
Affiliation(s)
- Yuanbiao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University; Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jungang Zhang
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kai Jiang
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Weiding Wu
- Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang; Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
29
|
Morales-Conde S, Licardie E, Alarcón I, Balla A. Indocyanine green (ICG) fluorescence guide for the use and indications in general surgery: recommendations based on the descriptive review of the literature and the analysis of experience. Cir Esp 2022; 100:534-554. [PMID: 35700889 DOI: 10.1016/j.cireng.2022.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/26/2021] [Indexed: 06/15/2023]
Abstract
Indocyanine Green is a fluorescent substance visible in near-infrared light. It is useful for the identification of anatomical structures (biliary tract, ureters, parathyroid, thoracic duct), the tissues vascularization (anastomosis in colorectal, esophageal, gastric, bariatric surgery, for plasties and flaps in abdominal wall surgery, liver resection, in strangulated hernias and in intestinal ischemia), for tumor identification (liver, pancreas, adrenal glands, implants of peritoneal carcinomatosis, retroperitoneal tumors and lymphomas) and sentinel node identification and lymphatic mapping in malignant tumors (stomach, breast, colon, rectum, esophagus and skin cancer). The evidence is very encouraging, although standardization of its use and randomized studies with higher number of patients are required to obtain definitive conclusions on its use in general surgery. The aim of this literature review is to provide a guide for the use of ICG fluorescence in general surgery procedures.
Collapse
Affiliation(s)
- Salvador Morales-Conde
- Unit of Innovation in Minimally Invasive Surgery, Department of General and Digestive Surgery, University Hospital Virgen del Rocio, University of Sevilla, Sevilla, Spain; Unit of General and Digestive Surgery, Hospital Quironsalud Sagrado Corazón, Sevilla, Spain.
| | - Eugenio Licardie
- Unit of General and Digestive Surgery, Hospital Quironsalud Sagrado Corazón, Sevilla, Spain.
| | - Isaias Alarcón
- Unit of Innovation in Minimally Invasive Surgery, Department of General and Digestive Surgery, University Hospital Virgen del Rocio, University of Sevilla, Sevilla, Spain.
| | - Andrea Balla
- Unit of Innovation in Minimally Invasive Surgery, Department of General and Digestive Surgery, University Hospital Virgen del Rocio, University of Sevilla, Sevilla, Spain; UOC of General and Minimally Invasive Surgery, Hospital "San Paolo", Civitavecchia, Rome, Italy.
| |
Collapse
|
30
|
Li Z, Li Z, Zaid W, Osborn ML, Li Y, Yao S, Xu J. Mouthwash as a non-invasive method of indocyanine green delivery for near-infrared fluorescence dental imaging. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-210326SSRR. [PMID: 35689334 PMCID: PMC9186466 DOI: 10.1117/1.jbo.27.6.066001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 05/23/2022] [Indexed: 05/25/2023]
Abstract
SIGNIFICANCE X-ray imaging serves as the mainstream imaging in dentistry, but it involves risk of ionizing radiation. AIM This study presents the feasibility of indocyanine green-assisted near-infrared fluorescence (ICG-NIRF) dental imaging with 785-nm NIR laser in the first (ICG-NIRF-I: 700 to 1000 nm) and second (ICG-NIRF-II: 1000 to 1700 nm) NIR wavelengths. APPROACH Sprague Dawley rats with different postnatal days were used as animal models. ICG, as a fluorescence agent, was delivered to dental structures by subcutaneous injection (SC) and oral administration (OA). RESULTS For SC method, erupted and unerupted molars could be observed from ICG-NIRF images at a short imaging time (<1 min). ICG-NIRF-II could achieve a better image contrast in unerupted molars at 24 h after ICG injection. The OA could serve as a non-invasive method for ICG delivery; it could also cause the glow-in-dark effect in unerupted molars. For erupted molars, OA can be considered as mouthwash and exhibits outstanding performance for delivery of ICG dye; erupted molar structures could be observed at a short imaging time (<1 min) and low ICG dose (0.05 mg / kg). CONCLUSIONS Overall, ICG-NIRF with mouthwash could perform in-vivo dental imaging in two NIR wavelengths at a short time and low ICG dose.
Collapse
Affiliation(s)
- Zhongqiang Li
- Louisiana State University, College of Engineering, Division of Electrical and Computer Engineering, Baton Rouge, Louisiana, United States
| | - Zheng Li
- Louisiana State University, College of Engineering, Division of Electrical and Computer Engineering, Baton Rouge, Louisiana, United States
| | - Waleed Zaid
- Louisiana State University Health Science Center, Oral and Maxillofacial Surgery, School of Dentistry, Baton Rouge, Louisiana, United States
| | - Michelle L. Osborn
- Louisiana State University, School of Veterinary Medicine, Department of Comparative Biomedical Science, Baton Rouge, Louisiana, United States
| | - Yanping Li
- University of Saskatchewan, School of Environment and Sustainability, Saskatoon, Saskatchewan, Canada
| | - Shaomian Yao
- Louisiana State University, School of Veterinary Medicine, Department of Comparative Biomedical Science, Baton Rouge, Louisiana, United States
| | - Jian Xu
- University of Saskatchewan, School of Environment and Sustainability, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
31
|
Azari F, Kennedy GT, Zhang K, Bernstein E, Maki RG, Gaughan C, Jarrar D, Pechet T, Kucharczuk J, Singhal S. Impact of Intraoperative Molecular Imaging after Fluorescent-Guided Pulmonary Metastasectomy for Sarcoma. J Am Coll Surg 2022; 234:748-758. [PMID: 35426386 PMCID: PMC9460458 DOI: 10.1097/xcs.0000000000000132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Intraoperative molecular imaging (IMI) has been shown to improve lesion detection during pulmonary sarcomatous metastasectomy. Our goal in this study was to evaluate whether data garnered from IMI-guided resection of pulmonary sarcoma metastasis translate to improved patient outcomes. STUDY DESIGN Fifty-two of 65 consecutive patients with a previous history of sarcomas found to have pulmonary nodules during screening were enrolled in a nonrandomized clinical trial. Patients underwent TumorGlow the day before surgery. Data on patient demographics, tumor biologic characteristics, preoperative assessment, and survival were included in the study analysis and compared with institutional historical data of patients who underwent metastasectomy without IMI. p values < 0.05 were considered significant. RESULTS IMI detected 42 additional lesions in 31 patients (59%) compared with the non-IMI cohort where 25% percent of patients had additional lesions detected using tactile and visual feedback only (p < 0.05). Median progression-free survival (PFS) for patients with IMI-guided pulmonary sarcoma metastasectomy was 36 months vs 28.6 months in the historical cohort (p < 0.05). IMI-guided pulmonary sarcoma metastasectomy had recurrence in the lung with a median time of 18 months compared with non-IMI group at 13 months (p < 0.05). Patients with synchronous lesions in the IMI group underwent systemic therapy at a statistically higher rate and tended to undergo routine screening at shorter interval. CONCLUSIONS IMI identifies a subset of sarcoma patients during pulmonary metastasectomy who have aggressive disease and informs the medical oncologist to pursue more aggressive systemic therapy. In this setting, IMI can serve both as a diagnostic and prognostic tool without conferring additional risk to the patient.
Collapse
Affiliation(s)
- Feredun Azari
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Gregory T Kennedy
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Kevin Zhang
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Elizabeth Bernstein
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Robert G Maki
- Hematology and Medical Oncology (Maki), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Colleen Gaughan
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Doraid Jarrar
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Taine Pechet
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - John Kucharczuk
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sunil Singhal
- From the Departments of Thoracic Surgery (Azari, Kennedy, Zhang, Bernstein, Gaughan, Jarrar, Pechet, Kucharczuk, Singhal), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
32
|
Ito R, Kamiya M, Urano Y. Molecular probes for fluorescence image-guided cancer surgery. Curr Opin Chem Biol 2022; 67:102112. [DOI: 10.1016/j.cbpa.2021.102112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/01/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022]
|
33
|
Whitlock RS, Patel KR, Yang T, Nguyen HN, Masand P, Vasudevan SA. Pathologic correlation with near infrared-indocyanine green guided surgery for pediatric liver cancer. J Pediatr Surg 2022; 57:700-710. [PMID: 34049689 DOI: 10.1016/j.jpedsurg.2021.04.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/06/2021] [Accepted: 04/18/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE Hepatoblastoma (HB) and hepatocellular carcinoma (HCC) are the most common primary malignant tumors of childhood. Intraoperative indocyanine green (ICG) administration with near-infrared imaging (NIR) has emerged as a surgical technology that can be used to assist with localization of pulmonary metastases secondary to HB; however, there has been limited application as an adjunct for resection of the primary liver tumor and assessment of extrahepatic disease. METHODS We present 14 patients treated for HB, HCC, and malignant rhabdoid tumor at our institution with the use of intraoperative NIR-ICG guidance. All patients were treated with 0.2-0.75 mg/kg IV ICG, 48-96 h prior to surgery. Intraoperative NIR-ICG guided imaging was performed with several commercial devices. RESULTS Intraoperative NIR-ICG guidance allowed pulmonary metastasectomy in five patients using thoracoscopy or thoracotomy allowing for visualization of multiple nodules not seen on preoperative imaging most of which were positive for malignancy. NIR-ICG guidance allowed for assessment of extrahepatic extension in three patients; an HCC patient with extrahepatic lymph node extension of disease, an HB patient with extrapulmonary thoracic recurrence in the diaphragm and chest wall, and a patient with tumor rupture at diagnosis with peritoneal nodules at the time of surgery. This technique was used to guide partial hepatectomy in 11 patients for which the technique enabled successful identification of tumor and tumor margins. Three patients had nonspecific staining of the liver secondary to decreased timing from ICG injection to surgery or biliary obstruction. NIR-ICG enabled resection of satellite HB lesions in three multifocal patients and confirmed a benign satellite lesion in two additional patients. CONCLUSIONS Intraoperative use of NIR-ICG imaging during partial hepatectomy enabled enhanced identification and guidance for surgical resection of extrahepatic disease and multifocal liver tumors for the treatment of children with primary liver cancer.
Collapse
Affiliation(s)
- Richard S Whitlock
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Surgical Oncology Program, Texas Children's Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX USA
| | - Kalyani R Patel
- Department of Pathology and Immunology, Texas Children's Hospital Liver Tumor Center, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX USA
| | - Tianyou Yang
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Surgical Oncology Program, Texas Children's Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX USA
| | - HaiThuy N Nguyen
- Singleton Department of Pediatric Radiology, Texas Children's Hospital Liver Tumor Program, Baylor College of Medicine, Houston, TX USA
| | - Prakash Masand
- Singleton Department of Pediatric Radiology, Texas Children's Hospital Liver Tumor Program, Baylor College of Medicine, Houston, TX USA
| | - Sanjeev A Vasudevan
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Texas Children's Surgical Oncology Program, Texas Children's Liver Tumor Program, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX USA.
| |
Collapse
|
34
|
Guía de uso e indicaciones de la fluorescencia con verde de indocianina (ICG) en cirugía general: recomendaciones basadas en la revisión descriptiva de la literatura y el análisis de la experiencia. Cir Esp 2022. [DOI: 10.1016/j.ciresp.2021.11.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
35
|
Sucher R, Scheuermann U, Rademacher S, Lederer A, Sucher E, Hau HM, Brandacher G, Schneeberger S, Gockel I, Seehofer D. Intraoperative reperfusion assessment of human pancreas allografts using hyperspectral imaging (HSI). Hepatobiliary Surg Nutr 2022; 11:67-77. [PMID: 35284501 PMCID: PMC8847868 DOI: 10.21037/hbsn-20-744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022]
Abstract
Background The most common causes of early graft loss in pancreas transplantation are insufficient blood supply and leakage of the intestinal anastomosis. Therefore, it is critical to monitor graft perfusion and oxygenation during the early post-transplant period. The goal of our pilot study was to evaluate the utility of hyperspectral imaging (HSI) in monitoring the microcirculation of the graft and adequate perfusion of the intestinal anastomosis during pancreatic allotransplantation. Methods We imaged pancreatic grafts and intestinal anastomosis in real-time in three consecutive, simultaneous pancreas-kidney transplantations using the TIVITA® HSI system. Further, the intraoperative oxygen saturation (StO2), tissue perfusion (near-infrared perfusion index, NIR), organ hemoglobin index (OHI), and tissue water index (TWI) were measured 15 minutes after reperfusion by HSI. Results All pancreas grafts showed a high and homogeneous StO2 (92.6%±10.45%). Intraoperative HSI analysis of the intestinal anastomosis displayed significant differences of StO2 (graft duodenum 67.46%±5.60% vs. recipient jejunum: 75.93%±4.71%, P<0.001) and TWI {graft duodenum: 0.63±0.09 [I (Index)] vs. recipient jejunum: 0.72±0.09 [I], P<0.001}. NIR and OHI did not display remarkable differences {NIR duodenum: 0.68±0.06 [I] vs. NIR jejunum: 0.69±0.04 [I], P=0.747; OHI duodenum: 0.70±0.12 [I] vs. OHI jejunum: 0.68±0.13 [I], P=0.449}. All 3 patients had an uneventful postoperative course with one displaying a Banff 1a rejection which was responsive to steroid treatment. Conclusions Our study shows that contact-free HSI has potential utility as a novel tool for real-time monitoring of human pancreatic grafts after reperfusion, which could improve the outcome of pancreas transplantation. Further investigations are required to determine the predictive value of intraoperative HSI imaging.
Collapse
Affiliation(s)
- Robert Sucher
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Uwe Scheuermann
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Sebastian Rademacher
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Andri Lederer
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Elisabeth Sucher
- Division of Hepatology, Clinic and Polyclinic for Gastroenterology, Hepatology, Infectiology, and Pneumology, University Hospital Leipzig, Leipzig, Germany
| | - Hans-Michael Hau
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany.,Department of Visceral, Transplantation, Vascular and Thoracic Surgery, University Hospital of Dresden, Dresden, Germany
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University, Baltimore, MD, USA
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Daniel Seehofer
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
36
|
Fluorescence Molecular Targeting of Colon Cancer to Visualize the Invisible. Cells 2022; 11:cells11020249. [PMID: 35053365 PMCID: PMC8773892 DOI: 10.3390/cells11020249] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/28/2021] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) is a common cause of cancer and cancer-related death. Surgery is the only curative modality. Fluorescence-enhanced visualization of CRC with targeted fluorescent probes that can delineate boundaries and target tumor-specific biomarkers can increase rates of curative resection. Approaches to enhancing visualization of the tumor-to-normal tissue interface are active areas of investigation. Nonspecific dyes are the most-used approach, but tumor-specific targeting agents are progressing in clinical trials. The present narrative review describes the principles of fluorescence targeting of CRC for diagnosis and fluorescence-guided surgery with molecular biomarkers for preclinical or clinical evaluation.
Collapse
|
37
|
Dolganova IN, Varvina DA, Shikunova IA, Alekseeva AI, Karalkin PA, Kuznetsov MR, Nikitin PV, Zotov AK, Mukhina EE, Katyba GM, Zaytsev KI, Tuchin VV, Kurlov VN. Proof of concept for the sapphire scalpel combining tissue dissection and optical diagnosis. Lasers Surg Med 2021; 54:611-622. [PMID: 34918347 DOI: 10.1002/lsm.23509] [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: 07/13/2021] [Revised: 10/18/2021] [Accepted: 11/27/2021] [Indexed: 11/08/2022]
Abstract
OBJECTIVES The development of compact diagnostic probes and instruments with an ability to direct access to organs and tissues and integration of these instruments into surgical workflows is an important task of modern physics and medicine. The need for such tools is essential for surgical oncology, where intraoperative visualization and demarcation of tumor margins define further prognosis and survival of patients. In this paper, the possible solution for this intraoperative imaging problem is proposed and its feasibility to detect tumorous tissue is studied experimentally. METHODS For this aim, the sapphire scalpel was developed and fabricated using the edge-defined film-fed growth technique aided by mechanical grinding, polishing, and chemical sharpening of the cutting edge. It possesses optical transparency, mechanical strength, chemical inertness, and thermal resistance alongside the presence of the as-grown hollow capillary channels in its volume for accommodating optical fibers. The rounding of the cutting edge exceeds the same for metal scalpels and can be as small as 110 nm. Thanks to these features, sapphire scalpel combines tissue dissection with light delivering and optical diagnosis. The feasibility for the tumor margin detection was studied, including both gelatin-based tissue phantoms and ex vivo freshly excised specimens of the basal cell carcinoma from humans and the glioma model 101.8 from rats. These tumors are commonly diagnosed either non-invasively or intraoperatively using different modalities of fluorescence spectroscopy and imaging, which makes them ideal candidates for our feasibility test. For this purpose, fiber-based spectroscopic measurements of the backscattered laser radiation and the fluorescence signals were carried out in the visible range. RESULTS Experimental studies show the feasibility of the proposed sapphire scalpel to provide a 2-mm-resolution of the tumor margins' detection, along with an ability to distinguish the tumor invasion region, which results from analysis of the backscattered optical fields and the endogenous or exogenous fluorescence data. CONCLUSIONS Our findings justified a strong potential of the sapphire scalpel for surgical oncology. However, further research and engineering efforts are required to optimize the sapphire scalpel geometry and the optical diagnosis protocols to meet the requirements of oncosurgery, including diagnosis and resection of neoplasms with different localizations and nosologies.
Collapse
Affiliation(s)
- Irina N Dolganova
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Bauman Moscow State Technical University, Moscow, Russia
| | - Daria A Varvina
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,International School "Medicine of the Future", Sechenov University, Moscow, Russia
| | - Irina A Shikunova
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka, Russia
| | - Anna I Alekseeva
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Research Institute of Human Morphology, Moscow, Russia
| | - Pavel A Karalkin
- Institute for Cluster Oncology, Sechenov University, Moscow, Russia.,Hertsen Moscow Oncology Research Institute, National Medical Research Radiological Centre, Moscow, Russia
| | | | - Pavel V Nikitin
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Arsen K Zotov
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka, Russia.,Bauman Moscow State Technical University, Moscow, Russia.,Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
| | | | - Gleb M Katyba
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka, Russia.,Bauman Moscow State Technical University, Moscow, Russia.,Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
| | - Kirill I Zaytsev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Bauman Moscow State Technical University, Moscow, Russia.,Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
| | - Valery V Tuchin
- Science Medical Center, Saratov State University, Saratov, Russia.,Institute of Precision Mechanics and Control of the Russian Academy of Sciences, Saratov, Russia.,National Research Tomsk University, Tomsk, Russia
| | - Vladimir N Kurlov
- Institute of Solid State Physics of the Russian Academy of Sciences, Chernogolovka, Russia.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.,Bauman Moscow State Technical University, Moscow, Russia
| |
Collapse
|
38
|
Sotiropoulou M, Mulita F, Verras GI, Schizas D, Papalampros A, Tchabashvili L, Kaplanis C, Liolis E, Perdikaris I, Maroulis I, Vailas M. A novel tool for visualization and detection of pancreatic neuroendocrine tumours. A 'fluorescent' world is calling for exploration? PRZEGLAD MENOPAUZALNY = MENOPAUSE REVIEW 2021; 20:207-210. [PMID: 35069073 PMCID: PMC8764961 DOI: 10.5114/pm.2021.110834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023]
Abstract
AIM OF THE STUDY Pancreatic neuroendocrine tumours (pNETs) are rare tumours with a propensity to metastasize. Physicians frequently face a huge clinical challenge during the localization of these lesions. The aim of this study is to investigate whether fluorescence-guided localization techniques with indocyanine green (ICG) can be utilized as a detection tool in pNETs, along with any other clinical implications of this technique. MATERIAL AND METHODS A thorough literature search in PubMed and Google Scholar, under the terms 'ICG OR Indocyanine OR Fluorescence AND Neuroendocrine' until 31 June 2021, regarding the utilization of indocyanine-fluorescence in localization of pancreatic neuroendocrine, was conducted by the authors, and the associated results are presented. RESULTS Indocyanine fluorescence imaging may facilitate the efforts of surgeons to identify occult pancreatic neuroendocrine lesions, assisting them in the identification of resection margins and delineation of the surgical anatomy when it is difficult to clarify. CONCLUSIONS Indocyanine-fluorescence imaging might play a pivotal role in pancreatic surgery in terms of localization for neuroendocrine tumours. However, further large-scale clinical studies are needed to assess the absolute indications and optimal use of this technique.
Collapse
Affiliation(s)
- Maria Sotiropoulou
- Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
| | - Francesk Mulita
- Department of Surgery, General University Hospital of Patras, Greece
| | | | - Dimitrios Schizas
- Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | - Elias Liolis
- Department of Internal Medicine, Division of Oncology, General University Hospital of Patras, Greece
| | | | - Ioannis Maroulis
- Department of Surgery, General University Hospital of Patras, Greece
| | - Michail Vailas
- Department of Surgery, General University Hospital of Patras, Greece
| |
Collapse
|
39
|
Li Z, Li Z, Ramos A, Boudreaux JP, Thiagarajan R, Mattison YB, Dunham ME, McWhorter AJ, Chen Q, Zhang J, Feng J, Li Y, Yao S, Xu J. Detection of pancreatic cancer by indocyanine green-assisted fluorescence imaging in the first and second near-infrared windows. Cancer Commun (Lond) 2021; 41:1431-1434. [PMID: 34787963 PMCID: PMC8696222 DOI: 10.1002/cac2.12236] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/22/2021] [Accepted: 10/28/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Zhongqiang Li
- Division of Electrical and Computer EngineeringCollege of Engineering, Louisiana State UniversityBaton RougeLA70803USA
| | - Zheng Li
- Division of Electrical and Computer EngineeringCollege of Engineering, Louisiana State UniversityBaton RougeLA70803USA
| | - Alexandra Ramos
- Department of Comparative Biomedical ScienceSchool of Veterinary Medicine, Louisiana State UniversityBaton RougeLA70803USA
| | - J. Philip Boudreaux
- Department of SurgerySchool of Medicine, Louisiana State University Health Science CenterNew OrleansLA70112USA
| | - Ramcharan Thiagarajan
- Department of SurgerySchool of Medicine, Louisiana State University Health Science CenterNew OrleansLA70112USA
| | - Yvette Bren Mattison
- Department of SurgerySchool of Medicine, Louisiana State University Health Science CenterNew OrleansLA70112USA
| | - Michael E. Dunham
- Department of OtolaryngologySchool of Medicine, Louisiana State University Health Science CenterNew OrleansLA70112USA
| | - Andrew J McWhorter
- Department of OtolaryngologySchool of Medicine, Louisiana State University Health Science CenterNew OrleansLA70112USA
| | - Qing Chen
- Division of Computer Science & EngineeringCollege of Engineering, Louisiana State UniversityBaton RougeLA70803USA
| | - Jian Zhang
- Division of Computer Science & EngineeringCollege of Engineering, Louisiana State UniversityBaton RougeLA70803USA
| | - Ji‐Ming Feng
- Department of Comparative Biomedical ScienceSchool of Veterinary Medicine, Louisiana State UniversityBaton RougeLA70803USA
| | - Yanping Li
- School of Environment and SustainabilityUniversity of SaskatchewanSaskatoonSKS7N 5C9Canada
| | - Shaomian Yao
- Department of Comparative Biomedical ScienceSchool of Veterinary Medicine, Louisiana State UniversityBaton RougeLA70803USA
| | - Jian Xu
- Division of Electrical and Computer EngineeringCollege of Engineering, Louisiana State UniversityBaton RougeLA70803USA
| |
Collapse
|
40
|
Wang L, Zhang D, Li J, Li F, Wei R, Jiang G, Xu H, Wang X, Zhou Y, Xi L. A novel ICG-labeled cyclic TMTP1 peptide dimer for sensitive tumor imaging and enhanced photothermal therapy in vivo. Eur J Med Chem 2021; 227:113935. [PMID: 34731764 DOI: 10.1016/j.ejmech.2021.113935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/26/2022]
Abstract
TMTP1 is a polypeptide independently screened in our laboratory, which can target tumors in situ and metastases. In previous work, we have successfully developed a near-infrared (NIR) probe TMTP1-PEG4-ICG for tumor imaging. However, the limited ability to target tumor micrometastases hinders its further clinical application. Multimerization of peptides has been extensively demonstrated as an effective strategy to increase receptor binding affinity due to "multivalent effect" or "apparent cooperative affinity". In this study, a novel TMTP1 homodimer-directed NIR probe (TMTP1-PEG4)2-ICG was successfully constructed and synthesized. The cyclic TMTP1 peptides were bridged by two PEG4 linkers and then labeled with ICG-NHS for tumor imaging and photothermal therapy. In vivo biodistribution were assessed in normal BALB/c mice, and tumor targeting abilities of (TMTP1-PEG4)2-ICG and its monomer were evaluated and compared in 4T1-bearing subcutaneous tumor and lymph node metastasis model mice. Biodistribution analysis in vivo revealed that (TMTP1-PEG4)2-ICG was cleared mainly in both liver and kidney dependent way. Comparing with free ICG dye or TMTP1-PEG4-ICG probe, this improved (TMTP1-PEG4)2-ICG dimer showed more sensitive tumor imaging and could clearly identify tumors at a minimum volume of 10 mm3. Additionally, when compared to its monomer, lymph node (LN) metastases could also be apparently visualized and easily distinguished from normal LN by the novel dimer at 24 h post-injection. The blocking study revealed that the tumor accumulation of this probe was specifically medicated by receptor-ligand interaction. Furthermore, with the increase in stability and tumor targeting ability of ICG in vivo, the probe could also be an attractive photothermal agent to significantly inhibit tumor growth under 808 nm NIR laser irradiation. In conclusion, our work revealed that the novel (TMTP1-PEG4)2-ICG dimer could be a promising theranostic agent for sensitive tumor imaging and imaging-guided photothermal therapy, indicating its broad prospects for further clinical transformation.
Collapse
Affiliation(s)
- Ling Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Danya Zhang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jie Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Fei Li
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Rui Wei
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Guiying Jiang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Hanjie Xu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xueqian Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ying Zhou
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Ling Xi
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
41
|
Schouw HM, Huisman LA, Janssen YF, Slart RHJA, Borra RJH, Willemsen ATM, Brouwers AH, van Dijl JM, Dierckx RA, van Dam GM, Szymanski W, Boersma HH, Kruijff S. Targeted optical fluorescence imaging: a meta-narrative review and future perspectives. Eur J Nucl Med Mol Imaging 2021; 48:4272-4292. [PMID: 34633509 PMCID: PMC8566445 DOI: 10.1007/s00259-021-05504-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/23/2021] [Indexed: 12/27/2022]
Abstract
Purpose The aim of this review is to give an overview of the current status of targeted optical fluorescence imaging in the field of oncology, cardiovascular, infectious and inflammatory diseases to further promote clinical translation. Methods A meta-narrative approach was taken to systematically describe the relevant literature. Consecutively, each field was assigned a developmental stage regarding the clinical implementation of optical fluorescence imaging. Results Optical fluorescence imaging is leaning towards clinical implementation in gastrointestinal and head and neck cancers, closely followed by pulmonary, neuro, breast and gynaecological oncology. In cardiovascular and infectious disease, optical imaging is in a less advanced/proof of concept stage. Conclusion Targeted optical fluorescence imaging is rapidly evolving and expanding into the clinic, especially in the field of oncology. However, the imaging modality still has to overcome some major challenges before it can be part of the standard of care in the clinic, such as the provision of pivotal trial data. Intensive multidisciplinary (pre-)clinical joined forces are essential to overcome the delivery of such compelling phase III registration trial data and subsequent regulatory approval and reimbursement hurdles to advance clinical implementation of targeted optical fluorescence imaging as part of standard practice. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05504-y.
Collapse
Affiliation(s)
- H M Schouw
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - L A Huisman
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Y F Janssen
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - R H J A Slart
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - R J H Borra
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Radiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - A T M Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - A H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - J M van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - R A Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Diagnostic Sciences, Ghent University Faculty of Medicine and Health Sciences, Gent, Belgium
| | - G M van Dam
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,AxelaRx/TRACER Europe BV, Groningen, The Netherlands
| | - W Szymanski
- Department of Radiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - H H Boersma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre of Groningen, Groningen, The Netherlands
| | - S Kruijff
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands. .,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.
| |
Collapse
|
42
|
Takahashi R, Ishizawa T, Sato M, Inagaki Y, Takanka M, Kuriki Y, Kamiya M, Ushiku T, Urano Y, Hasegawa K. Fluorescence Imaging Using Enzyme-Activatable Probes for Real-Time Identification of Pancreatic Cancer. Front Oncol 2021; 11:714527. [PMID: 34490111 PMCID: PMC8417470 DOI: 10.3389/fonc.2021.714527] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 01/11/2023] Open
Abstract
Introduction Radical resection is the only curative treatment for pancreatic cancer, which is a life-threatening disease. However, it is often not easy to accurately identify the extent of the tumor before and during surgery. Here we describe the development of a novel method to detect pancreatic tumors using a tumor-specific enzyme-activatable fluorescence probe. Methods Tumor and non-tumor lysate or small specimen collected from the resected specimen were selected to serve as the most appropriate fluorescence probe to distinguish cancer tissues from noncancerous tissues. The selected probe was sprayed onto the cut surface of the resected specimen of cancer tissue to acquire a fluorescence image. Next, we evaluated the ability of the probe to detect the tumor and calculated the tumor-to-background ratio (TBR) by comparing the fluorescence image with the pathological extent of the tumor. Finally, we searched for a tumor-specific enzyme that optimally activates the selected probe. Results Using a library comprising 309 unique fluorescence probes, we selected GP-HMRG as the most appropriate activatable fluorescence probe. We obtained eight fluorescence images of resected specimens, among which four approximated the pathological findings of the tumor, which achieved the highest TBR. Finally, dipeptidyl-peptidase IV (DPP-IV) or a DPP-IV-like enzyme was identified as the target enzyme. Conclusion This novel method may enable rapid and real-time visualization of pancreatic cancer through the enzymatic activities of cancer tissues.
Collapse
Affiliation(s)
- Ryugen Takahashi
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Takeaki Ishizawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Masumitsu Sato
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yoshinori Inagaki
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Mariko Takanka
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yugo Kuriki
- Laboratory of Chemistry and Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Mako Kamiya
- Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuteru Urano
- Laboratory of Chemistry and Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
43
|
Li D, Yang M, Liang M, Mei C, Lin Y, Yang F, Xiao Y, Chen Y, Wang F, Mao J, Su Z. c-Met-targeted near-infrared fluorescent probe for real-time depiction and dissection of perineural invasion and lymph node metastasis lesions in pancreatic ductal adenocarcinoma xenograft models. Biomater Sci 2021; 9:6737-6752. [PMID: 34254599 DOI: 10.1039/d1bm00674f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a fatal malignant tumour, has a high postoperative recurrence rate, mainly due to the difficulty of discerning occult lesions, including those related to perineural invasion (PNI) and lymph node metastasis (LNM). Cellular mesenchymal-epithelial transition factor (c-Met), an excellent imaging marker, is aberrantly expressed in the majority of PDACs. Thus, we plan to utilize a c-Met-targeted near-infrared fluorescent (NIRF) probe for real-time visualization and dissection of PDAC, and corresponding PNI and LNM lesions. Immunohistochemistry showed c-Met expression in PDAC, PNI and LNM reached 94.3% (100/106), 88.3% (53/60), and 71.4% (25/35), respectively, and its expression in PNI and LNM was significantly correlated with that in primary PDAC (r = 0.66, p < 0.0001 and r = 0.44, p < 0.01, respectively). SHRmAb-IR800 was successfully synthesized using an anti-c-Met antibody and a NIRF dye. The in vitro targeting ability of SHRmAb-IR800 was higher in CFPAC1 cells (c-Met positive) than in Miapaca-2 cells (c-Met negative) (p < 0.05). In vivo NIRF imaging of CFPAC1 subcutaneous tumours demonstrated higher accumulation of SHRmAb-IR800 than the control probe (p < 0.05). The signal-to-background ratio (TBR) of an orthotopic PDAC tumour was 3.38 ± 0.46, and imaging with SHRmAb-IR800 facilitated the resection of metastatic lesions with sensitivity and specificity values of 93.3% (56/60) and 87.1% (27/31), respectively. Furthermore, tiny PNI and LNM lesions in xenograft models were detected by NIRF imaging, with TBRs measuring 2.59 ± 0.19 and 2.88 ± 0.72, respectively. Therefore, the clinical translation of this probe might shed new light on NIRF-guided pancreatectomy and improve the surgical prognosis of PDAC patients.
Collapse
Affiliation(s)
- Dan Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Meilin Yang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Mingzhu Liang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China and Center for Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China. and Department of Radiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Chaoming Mei
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yujing Lin
- Department of Pathology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Fan Yang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yitai Xiao
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yuechuan Chen
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Fen Wang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province 510080, China.
| | - Junjie Mao
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China and Center for Interventional Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| | - Zhongzhen Su
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China and Department of Ultrasound, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| |
Collapse
|
44
|
Yang L, Park J, Marjanovic M, Chaney EJ, Spillman DR, Phillips H, Boppart SA. Intraoperative Label-Free Multimodal Nonlinear Optical Imaging for Point-of-Procedure Cancer Diagnostics. IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS : A PUBLICATION OF THE IEEE LASERS AND ELECTRO-OPTICS SOCIETY 2021; 27:6801412. [PMID: 33746497 PMCID: PMC7978401 DOI: 10.1109/jstqe.2021.3054578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Intraoperative imaging in surgical oncology can provide information about the tumor microenvironment as well as information about the tumor margin. Visualizing microstructural features and molecular and functional dynamics may provide important diagnostic and prognostic information, especially when obtained in real-time at the point-of-procedure. A majority of current intraoperative optical techniques are based on the use of the labels, such as fluorescent dyes. However, these exogenous agents disrupt the natural microenvironment, perturb biological processes, and alter the endogenous optical signatures that cells and the microenvironment can provide. Portable nonlinear imaging systems have enabled intraoperative imaging for real-time detection and diagnosis of tissue. We review the development of a label-free multimodal nonlinear optical imaging technique that was adapted into a portable imaging system for intraoperative optical assessment of resected human breast tissue. New developments have applied this technology to assessing needle-biopsy specimens. Needle-biopsy procedures most always precede surgical resection and serve as the first sampling of suspicious masses for diagnosis. We demonstrate the diagnostic feasibility of imaging core needle-biopsy specimens during veterinary cancer surgeries. This intraoperative label-free multimodal nonlinear optical imaging technique can potentially provide a powerful tool to assist in cancer diagnosis at the point-of-procedure.
Collapse
Affiliation(s)
| | | | | | | | - Darold R Spillman
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Heidi Phillips
- Small Animal Surgery, Veterinary Teaching Hospital, University of Illinois College of Veterinary Medicine, Urbana, IL 61802 USA
| | - Stephen A Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| |
Collapse
|
45
|
Azari F, Kennedy G, Bernstein E, Hadjipanayis C, Vahrmeijer AL, Smith BL, Rosenthal E, Sumer B, Tian J, Henderson ER, Lee A, Nguyen Q, Gibbs SL, Pogue BW, Orringer DA, Charalampaki P, Martin LW, Tanyi JL, Kenneth Lee M, Lee JYK, Singhal S. Intraoperative molecular imaging clinical trials: a review of 2020 conference proceedings. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-210050VR. [PMID: 34002555 PMCID: PMC8126806 DOI: 10.1117/1.jbo.26.5.050901] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/28/2021] [Indexed: 05/15/2023]
Abstract
SIGNIFICANCE Surgery is often paramount in the management of many solid organ malignancies because optimal resection is a major factor in disease-specific survival. Cancer surgery has multiple challenges including localizing small lesions, ensuring negative surgical margins around a tumor, adequately staging patients by discriminating positive lymph nodes, and identifying potential synchronous cancers. Intraoperative molecular imaging (IMI) is an emerging potential tool proposed to address these issues. IMI is the process of injecting patients with fluorescent-targeted contrast agents that highlight cancer cells prior to surgery. Over the last 5 to 7 years, enormous progress has been achieved in tracer development, near-infrared camera approvals, and clinical trials. Therefore, a second biennial conference was organized at the University of Pennsylvania to gather surgical oncologists, scientists, and experts to discuss new investigative findings in the field. Our review summarizes the discussions from the conference and highlights findings in various clinical and scientific trials. AIM Recent advances in IMI were presented, and the importance of each clinical trial for surgical oncology was critically assessed. A major focus was to elaborate on the clinical endpoints that were being utilized in IMI trials to advance the respective surgical subspecialties. APPROACH Principal investigators presenting at the Perelman School of Medicine Abramson Cancer Center's second clinical trials update on IMI were selected to discuss their clinical trials and endpoints. RESULTS Multiple phase III, II, and I trials were discussed during the conference. Since the approval of 5-ALA for commercial use in neurosurgical malignancies, multiple tracers and devices have been developed to address common challenges faced by cancer surgeons across numerous specialties. Discussants also presented tracers that are being developed for delineation of normal anatomic structures that can serve as an adjunct during surgical procedures. CONCLUSIONS IMI is increasingly being recognized as an improvement to standard oncologic surgical resections and will likely advance the art of cancer surgery in the coming years. The endpoints in each individual surgical subspecialty are varied depending on how IMI helps each specialty solve their clinical challenges.
Collapse
Affiliation(s)
- Feredun Azari
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Gregory Kennedy
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Elizabeth Bernstein
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | | | | | - Barbara L. Smith
- Harvard University, School of Medicine, Boston, Massachusetts, United States
| | - Eben Rosenthal
- Stanford University, School of Medicine, Stanford, California, United States
| | - Baran Sumer
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Jie Tian
- Chinese Academy of Sciences/Institute of Automation, Beijing, China
| | - Eric R. Henderson
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
| | - Amy Lee
- University of Washington, School of Medicine, Seattle, Washington, United States
| | - Quyen Nguyen
- University of California San Diego, School of Medicine, San Diego, California, United States
| | - Summer L. Gibbs
- Oregon Health & Science University, Knight Cancer Institute, School of Medicine, Portland, Oregon, United States
| | - Brian W. Pogue
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
- Thayer School of Engineering at Dartmouth, Hanover, New Hampshire, United States
| | | | | | - Linda W. Martin
- University of Virginia, School of Medicine, Charlottesville, Virginia, United States
| | - Janos L. Tanyi
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Major Kenneth Lee
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - John Y. K. Lee
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
| | - Sunil Singhal
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States
- Address all correspondence to Sunil Singhal,
| |
Collapse
|
46
|
Iyer SG. Fluorescence imaging in hepatobiliary surgery - hope and hype. Singapore Med J 2021; 62:157-158. [PMID: 33948666 PMCID: PMC8801828 DOI: 10.11622/smedj.2021035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Shridhar Ganpathi Iyer
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Singapore
- Liver Transplant Programme, National University Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
47
|
Abraham T, McGovern CO, Linton SS, Wilczynski Z, Adair JH, Matters GL. Aptamer-Targeted Calcium Phosphosilicate Nanoparticles for Effective Imaging of Pancreatic and Prostate Cancer. Int J Nanomedicine 2021; 16:2297-2309. [PMID: 33776434 PMCID: PMC7989532 DOI: 10.2147/ijn.s295740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/06/2021] [Indexed: 01/22/2023] Open
Abstract
Purpose Accurate tumor identification and staging can be difficult. Aptamer-targeted indocyanine green (ICG)-nanoparticles can enhance near-infrared fluorescent imaging of pancreatic and prostate tumors and could improve early cancer detection. This project explored whether calcium-phosphosilicate nanoparticles, also known as NanoJackets (NJs), that were bioconjugated with a tumor-specific targeting DNA aptamer could improve the non-invasive detection of pancreatic and prostate tumors. Methods Using in vivo near-infrared optical imaging and ex vivo fluorescence analysis, DNA aptamer-targeted ICG-loaded NJs were compared to untargeted NJs for detection of tumors. Results Nanoparticles were bioconjugated with the DNA aptamer AP1153, which binds to the CCK-B receptor (CCKBR). Aptamer bioconjugated NJs were not significantly increased in size compared with unconjugated nanoparticles. AP1153-ICG-NJ accumulation in orthotopic pancreatic tumors peaked at 18 h post-injection and the ICG signal was cleared by 36 h with no evidence on uptake by non-tumor tissues. Ex vivo tumor imaging confirmed the aptamer-targeted NJs accumulated to higher levels than untargeted NJs, were not taken up by normal pancreas, exited from the tumor vasculature, and were well-dispersed throughout pancreatic and prostate tumors despite extensive fibrosis. Specificity for AP1153-NJ binding to the CCK-B receptor on pancreatic tumor cells was confirmed by pre-treating tumor-bearing mice with the CCK receptor antagonist proglumide. Proglumide pre-treatment reduced the in vivo tumoral accumulation of AP1153-NJs to levels comparable to that of untargeted NJs. Conclusion Through specific interactions with CCK-B receptors, tumor-targeted nanoparticles containing either ICG or rhodamine WT were well distributed throughout the matrix of both pancreatic and prostate tumors. Tumor-targeted NJs carrying various imaging agents can enhance tumor detection.
Collapse
Affiliation(s)
- Thomas Abraham
- Departments of Neural and Behavioral Sciences and the Microscopy Imaging Core Facility, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Christopher O McGovern
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Samuel S Linton
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Zachary Wilczynski
- Departments of Materials Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - James H Adair
- Departments of Materials Science, The Pennsylvania State University, University Park, PA, 16802, USA.,Department of Biomedical Engineering and Pharmacology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Gail L Matters
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
48
|
Bagaria SP, Gronchi A. Distal Pancreatectomy for Primary Retroperitoneal Sarcoma-Clinical Implications and Future Directions. Ann Surg Oncol 2021; 28:6890-6891. [PMID: 33638045 DOI: 10.1245/s10434-021-09766-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 11/18/2022]
Affiliation(s)
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
49
|
Unique Benefits of Tumor-Specific Nanobodies for Fluorescence Guided Surgery. Biomolecules 2021; 11:biom11020311. [PMID: 33670740 PMCID: PMC7921980 DOI: 10.3390/biom11020311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor-specific fluorescence labeling is promising for real-time visualization of solid malignancies during surgery. There are a number of technologies to confer tumor-specific fluorescence. Antibodies have traditionally been used due to their versatility in modifications; however, their large size hampers efficient fluorophore delivery. Nanobodies are a novel class of molecules, derived from camelid heavy-chain only antibodies, that have shown promise for tumor-specific fluorescence labeling. Nanobodies are ten times smaller than standard antibodies, while maintaining antigen-binding capacity and have advantageous features, including rapidity of tumor labeling, that are reviewed in the present report. The present report reviews special considerations needed in developing nanobody probes, the status of current literature on the use of nanobody probes in fluorescence guided surgery, and potential challenges to be addressed for clinical translation.
Collapse
|
50
|
Goldstein SD, Heaton TE, Bondoc A, Dasgupta R, Abdelhafeez A, Davidoff AM, Lautz TB. Evolving applications of fluorescence guided surgery in pediatric surgical oncology: A practical guide for surgeons. J Pediatr Surg 2021; 56:215-223. [PMID: 33189300 DOI: 10.1016/j.jpedsurg.2020.10.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/09/2020] [Accepted: 10/06/2020] [Indexed: 01/10/2023]
Abstract
Fluorescence-guided surgery (FGS) is an increasingly available and popular method of visual field augmentation. The basic premise of FGS entails injection of fluorescent indocyanine green (ICG) and subsequent detection with a near-infrared (NIR) camera. For pediatric surgical oncologists, FGS remains experimental but is a promising modality for identifying tumor margins, locating metastases, performing sentinel lymph node biopsies, protecting peritumoral structures of interest, and facilitating reconstruction. Familiarity with basic ICG pharmacokinetics and NIR detection optics is critical for surgeons wishing to judiciously use FGS, as its success is firmly grounded in a thorough understanding of its capabilities and limitations. In this practical guide, we outline several well-described and innovative FGS applications by disease type, including their methods of administration, modes of detection, and typical ICG dosing paradigms. LEVEL OF EVIDENCE: V.
Collapse
Affiliation(s)
- Seth D Goldstein
- Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Todd E Heaton
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander Bondoc
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Roshni Dasgupta
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN
| | - Timothy B Lautz
- Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL.
| |
Collapse
|