1
|
Liu Y, Valji K, Monsky W, Zheng C, Yang X. Optical imaging guidance in oncologic surgery and interventional oncology. Pharmacol Res 2025; 212:107612. [PMID: 39826822 DOI: 10.1016/j.phrs.2025.107612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Over recent decades, optical imaging (OI) has become an integral part of medical imaging, offering significant advantages over other modalities, such as computed tomography (CT) and magnetic resonance imaging (MRI). OI is distinguished by its real-time imaging capability, cost-effectiveness, portability, absence of ionizing radiation, and high patient acceptability. The introduction of advanced optical dyes (including FDA-approved agents like indocyanine green, Cytalux, and Gleolan) has greatly enhanced its clinical utility. OI has shown clear benefits in the management of patients with cancer, originally by open surgery and now extending to minimally invasive, image-guided interventional procedures. This review highlights recent developments in OI for oncology, emphasizing its benefits for clinicians in guiding surgical and interventional procedures.
Collapse
Affiliation(s)
- Yiming Liu
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA; Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Karim Valji
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA
| | - Wayne Monsky
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoming Yang
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA.
| |
Collapse
|
2
|
Kotb A, Hafeji Z, Jesry F, Lintern N, Pathak S, Smith AM, Lutchman KRD, de Bruin DM, Hurks R, Heger M, Khaled YS. Intra-Operative Tumour Detection and Staging in Pancreatic Cancer Surgery: An Integrative Review of Current Standards and Future Directions. Cancers (Basel) 2024; 16:3803. [PMID: 39594758 PMCID: PMC11592681 DOI: 10.3390/cancers16223803] [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: 06/09/2024] [Revised: 10/15/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Surgical resection for pancreatic ductal adenocarcinoma (PDAC) entails the excision of the primary tumour and regional lymphadenectomy. This traditional strategy is challenged by the high rate of early recurrence, suggesting inadequate disease staging. Novel methods of intra-operative staging are needed to allow surgical resection to be tailored to the disease's biology. METHODS A search of published articles on the PubMed and Embase databases was performed using the terms 'pancreas' OR 'pancreatic' AND 'intra-operative staging/detection' OR 'guided surgery'. Articles published between January 2000 and June 2023 were included. Technologies that offered intra-operative staging and tailored treatment were curated and summarised in the following integrative review. RESULTS lymph node (LN) mapping and radioimmunoguided surgery have shown promising results but lacked practicality to facilitate real-time intra-operative staging for PDAC. Fluorescence-guided surgery (FGS) offers high contrast and sensitivity, enabling the identification of cancerous tissue and positive LNs with improved precision following intravenous administration of a fluorescent agent. The unique properties of optical coherence tomography and ultrasound elastography lend themselves to be platforms for virtual biopsy intra-operatively. CONCLUSIONS Accurate intra-operative staging of PDAC, localisation of metastatic LNs, and identification of extra-pancreatic disease remain clinically unmet needs under current detection methods and staging standards. Tumour-specific FGS combined with other diagnostic and therapeutic modalities could improve tumour detection and staging in patients with PDAC.
Collapse
Affiliation(s)
- Ahmed Kotb
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Zaynab Hafeji
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Fadel Jesry
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Nicole Lintern
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
| | - Samir Pathak
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| | - Andrew M. Smith
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| | - Kishan R. D. Lutchman
- Department of Surgery, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Daniel M. de Bruin
- Department of Biomedical Engineering and Physics, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Rob Hurks
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Yazan S. Khaled
- Leeds Institute of Medical Research, University of Leeds, Leeds LS2 9JT, UK
- The Pancreato-Biliary Unit, St James’s University Teaching Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
3
|
Yao S, Wang Y, Mou X, Yang X, Cai Y. Recent advances of photoresponsive nanomaterials for diagnosis and treatment of acute kidney injury. J Nanobiotechnology 2024; 22:676. [PMID: 39501286 PMCID: PMC11536863 DOI: 10.1186/s12951-024-02906-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/04/2024] [Indexed: 11/09/2024] Open
Abstract
Non-invasive imaging in the near-infrared region (NIR) offers enhanced tissue penetration, reduced spontaneous fluorescence of biological tissues, and improved signal-to-noise ratio (SNR), rendering it more suitable for in vivo deep tissue imaging. In recent years, a plethora of NIR photoresponsive materials have been employed for disease diagnosis, particularly acute kidney injury (AKI). These encompass inorganic nonmetallic materials such as carbon (C), silicon (Si), phosphorus (P), and upconversion nanoparticles (UCNPs); precious metal nanoparticles like gold and silver; as well as small molecule and organic semiconductor polymer nanoparticles with near infrared responsiveness. These materials enable effective therapy triggered by NIR light and serve as valuable tools for monitoring AKI in living systems. The review provides a concise overview of the current state and pathological characteristics of AKI, followed by an exploration of the application of nanomaterials and photoresponsive nanomaterials in AKI. Finally, it presents the design challenges and prospects associated with NIR photoresponsive materials in AKI.
Collapse
Affiliation(s)
- Shijie Yao
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Yinan Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Xianghong Yang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Yu Cai
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- Clinical Research Institute, Zhejiang Provincial People's Hospital, (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
4
|
Cheng H, Xu H, Peng B, Huang X, Hu Y, Zheng C, Zhang Z. Illuminating the future of precision cancer surgery with fluorescence imaging and artificial intelligence convergence. NPJ Precis Oncol 2024; 8:196. [PMID: 39251820 PMCID: PMC11385925 DOI: 10.1038/s41698-024-00699-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Real-time and accurate guidance for tumor resection has long been anticipated by surgeons. In the past decade, the flourishing material science has made impressive progress in near-infrared fluorophores that may fulfill this purpose. Fluorescence imaging-guided surgery shows great promise for clinical application and has undergone widespread evaluations, though it still requires continuous improvements to transition this technique from bench to bedside. Concurrently, the rapid progress of artificial intelligence (AI) has revolutionized medicine, aiding in the screening, diagnosis, and treatment of human doctors. Incorporating AI helps enhance fluorescence imaging and is poised to bring major innovations to surgical guidance, thereby realizing precision cancer surgery. This review provides an overview of the principles and clinical evaluations of fluorescence-guided surgery. Furthermore, recent endeavors to synergize AI with fluorescence imaging were presented, and the benefits of this interdisciplinary convergence were discussed. Finally, several implementation strategies to overcome technical hurdles were proposed to encourage and inspire future research to expedite the clinical application of these revolutionary technologies.
Collapse
Affiliation(s)
- Han Cheng
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China
| | - Hongtao Xu
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China
| | - Boyang Peng
- School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | - Xiaojuan Huang
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China
| | - Yongjie Hu
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China
| | - Chongyang Zheng
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China.
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China.
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China.
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China.
| | - Zhiyuan Zhang
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China.
- College of Stomatology, Shanghai Jiao Tong University & National Center for Stomatology, Shanghai, 200011, P. R. China.
- National Clinical Research Center for Oral Diseases & Shanghai Key Laboratory of Stomatology, Shanghai, 200011, P. R. China.
- Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China.
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, P. R. China.
| |
Collapse
|
5
|
White HW, Naveed AB, Campbell BR, Lee YJ, Baik FM, Topf M, Rosenthal EL, Hom ME. Infrared Fluorescence-guided Surgery for Tumor and Metastatic Lymph Node Detection in Head and Neck Cancer. Radiol Imaging Cancer 2024; 6:e230178. [PMID: 38940689 PMCID: PMC11287229 DOI: 10.1148/rycan.230178] [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: 10/09/2023] [Revised: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024]
Abstract
In patients with head and neck cancer (HNC), surgical removal of cancerous tissue presents the best overall survival rate. However, failure to obtain negative margins during resection has remained a steady concern over the past 3 decades. The need for improved tumor removal and margin assessment presents an ongoing concern for the field. While near-infrared agents have long been used in imaging, investigation of these agents for use in HNC imaging has dramatically expanded in the past decade. Targeted tracers for use in primary and metastatic lymph node detection are of particular interest, with panitumumab-IRDye800 as a major candidate in current studies. This review aims to provide an overview of intraoperative near-infrared fluorescence-guided surgery techniques used in the clinical detection of malignant tissue and sentinel lymph nodes in HNC, highlighting current applications, limitations, and future directions for use of this technology within the field. Keywords: Molecular Imaging-Cancer, Fluorescence © RSNA, 2024.
Collapse
Affiliation(s)
- Haley W. White
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Abdullah Bin Naveed
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Benjamin R. Campbell
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Yu-Jin Lee
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Fred M. Baik
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Michael Topf
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Eben L. Rosenthal
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| | - Marisa E. Hom
- From the University of Michigan School of Medicine, Ann Arbor, Mich
(H.W.W.); Department of Otolaryngology-Head and Neck Surgery, Vanderbilt
University Medical Center, 2220 Pierce Ave, PRB 754, Nashville, TN 37232
(A.B.N., B.R.C., M.T., E.L.R., M.E.H.); and Department of Otolaryngology-Head
and Neck Surgery, Stanford University School of Medicine, Stanford, Calif
(Y.J.L., F.M.B.)
| |
Collapse
|
6
|
Kamimura T. Blood Flow in the Meniscus Can Be Visualized Arthroscopically Using an Intravenous Indocyanine Green Solution Diluted 10× in a Pig Model. Arthrosc Sports Med Rehabil 2024; 6:100932. [PMID: 39006800 PMCID: PMC11240037 DOI: 10.1016/j.asmr.2024.100932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/17/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose To determine the optimal indocyanine green (ICG) dose required for assessing vascularity in the meniscus using ICG fluorescence-guided knee arthroscopy in a pig model. Methods A 3-month-old Japanese esculent female pig was used in this study. Intravenous injections of ICG (25 mg) were administered with 2.0 mL of 5×, 10×, 100×, and 1,000× diluted solutions. An additional experiment was conducted to assess the microvasculature within the meniscus considering the results of the optimal dilution setting. A radial tear was purposely induced in the middle-to-posterior section of the medial meniscus to observe vascularity in the cross-sectioned meniscus; the optimal ICG dilution was administered. Results No fluorescence was detected in the meniscus with solutions diluted by 1,000× and 100×. Fluorescence was visualized at the anterior portion of the synovium and the anterior cruciate ligament using ICG diluted by 10×. Diluting ICG by 5×, contrast enhancement was too intense for observation. Therefore, the 10× diluted solution was considered the optimal setting for knee arthroscopy and observation of the radial tear. No fluorescence was observed in the cross section of the medial meniscus. Arterial hemorrhage was observed by stimulating the fluorescence-dyed synovium adjacent to the tear site. Through the additional waiting time after stimulating the tear site, the hemorrhage inside the meniscus became more intense. Conclusions The optimal dilution and dose setting of ICG for knee arthroscopy was 10× in a 2.0-mL intravenous injection. The meniscus showed no active blood flow, even in the red-red zone. This finding might support the notion that blood flow cannot be initiated, without synovial stimulation, even in vascular areas. Clinical Relevance This study could determine an ICG solution suitable for ICG fluorescence-guided knee arthroscopy. This finding could be valuable in future research focusing on case-specific meniscal vascularization under arthroscopy, particularly applying these findings to human meniscal treatment.
Collapse
Affiliation(s)
- Tamiko Kamimura
- Department of Orthopaedic Surgery, Tokorozawa Chuo Hospital, Tokorozawa, Japan
| |
Collapse
|
7
|
Wang M, Xie H, Tang BZ, Wang WX. Novel Near-Infrared-II In Vivo Visualization Revealed Rapid Calcium Intestine Turnover in Daphnia magna with Delayed Impact by Cadmium and Acidification. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:4558-4570. [PMID: 38408313 DOI: 10.1021/acs.est.3c10468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Calcium is a highly demanded metal, and its transport across the intestine of Daphnia magna remains a significant unresolved question. Due to technical constraints, the visualization of the kinetic process of Ca passage through D. magna has been challenging. Here, we developed the second near-infrared Ca sensor (NIR-II Ca) and conducted real-time in vivo imaging of Ca in daphnids with a high signal-to-noise ratio, deep tissue penetration, and minimal damage. Through the utilization of the NIR-II Ca sensor, we for the first time visualized and quantified the kinetic process of Ca passage in the intestine in real time. The results revealed that trophically available Ca passed through the intestines in 24 h, whereas waterborne Ca required only 35 min. This rapid "flushing through" mechanism established waterborne Ca as the primary source of Ca absorption. However, environmental stressors such as water acidification and cadmium significantly delayed the Ca passage and absorption. The development of NIR imaging and sensors allows for real-time dynamic visualization of contaminants/nutrients in organisms and holds great potential as a powerful tool for future studies into material kinetic processes in living animals.
Collapse
Affiliation(s)
- Mengyu Wang
- School of Energy and Environment, State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China
- Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Huilin Xie
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Ben Zhong Tang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong 999077, China
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| | - Wen-Xiong Wang
- School of Energy and Environment, State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China
- Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
8
|
Colazo JM, Prasad K, Miller A, Sharif K, Aweeda M, Fassler C, Singh R, Schwartz HS, Lawrenz JM, Holt GE, Topf MC. 3D Specimen Scanning and Mapping in Musculoskeletal Oncology: A Feasibility Study. Ann Surg Oncol 2024; 31:2051-2060. [PMID: 38133863 DOI: 10.1245/s10434-023-14757-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/25/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Surgical resection is the primary treatment for bone and soft tissue tumors. Negative margin status is a key factor in prognosis. Given the three-dimensional (3D) anatomic complexity of musculoskeletal tumor specimens, communication of margin results between surgeons and pathologists is challenging. We sought to perform ex vivo 3D scanning of musculoskeletal oncology specimens to enhance communication between surgeons and pathologists. METHODS Immediately after surgical resection, 3D scanning of the fresh specimen is performed prior to frozen section analysis. During pathologic grossing, whether frozen or permanent, margin sampling sites are annotated on the virtual 3D model using computer-aided design (CAD) software. RESULTS 3D scanning was performed in seven cases (six soft tissue, one bone), with specimen mapping on six cases. Intraoperative 3D scanning and mapping was performed in one case in which the location of margin sampling was shown virtually in real-time to the operating surgeon to help achieve a negative margin. In six cases, the 3D model was used to communicate final permanent section analysis. Soft tissue, cartilage, and bone (including lytic lesions within bone) showed acceptable resolution. CONCLUSIONS Virtual 3D scanning and specimen mapping is feasible and may allow for enhanced documentation and communication. This protocol provides useful information for anatomically complex musculoskeletal tumor specimens. Future studies will evaluate the effect of the protocol on positive margin rates, likelihood that a re-resection contains additional malignancy, and exploration of targeted adjuvant radiation protocols using a patient-specific 3D specimen map.
Collapse
Affiliation(s)
- Juan M Colazo
- School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Kavita Prasad
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexis Miller
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kayvon Sharif
- School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Marina Aweeda
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carly Fassler
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Reena Singh
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Herbert S Schwartz
- Division of Musculoskeletal Oncology, Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua M Lawrenz
- Division of Musculoskeletal Oncology, Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ginger E Holt
- Division of Musculoskeletal Oncology, Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael C Topf
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
9
|
Brollo PP, Bresadola V. Enhancing visualization and guidance in general surgery: a comprehensive and narrative review of the current cutting-edge technologies and future perspectives. J Gastrointest Surg 2024; 28:179-185. [PMID: 38445941 DOI: 10.1016/j.gassur.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/25/2023] [Accepted: 12/08/2023] [Indexed: 03/07/2024]
Abstract
BACKGROUND In the last decade, there has been a great effort in developing new technologies to enhance surgical visualization and guidance. This comprehensive and narrative review aimed to provide a wide and extensive overview of the current state of the art on this topic and their near-future perspectives linked to the development of artificial intelligence (AI), by focusing on the most recent and relevant literature. METHODS A comprehensive and narrative review of the literature was performed by searching specific terms on PubMed/MEDLINE, Scopus, and Embase databases, assessing the current state of the art on this topic. RESULTS Fluorescence-guided surgery, contrast-enhanced ultrasound (CEUS), ultra-high frequency ultrasound (UHFUS), photoacoustic imaging (PAI), and augmented reality (AR) are boosting the field of image-guided techniques as the rapid development of AI in surgery is promising a more automated decision-making and surgical movements in the operating room. CONCLUSION Fluorescence-guided surgery, CEUS, UHFUS, PAI, and AR are becoming crucial to give surgeons a new level of information during the intervention, with the right timing and sequence, and represent the future of surgery. As many more controlled studies are needed to validate the employment of these technologies, the next generation of surgeons must become more familiar with the basics of AI to better incorporate new tools into the daily surgical practice of the future.
Collapse
Affiliation(s)
- Pier Paolo Brollo
- Department of Medicine, General Surgery Department and Simulation Center, Academic Hospital of Udine, University of Udine, Udine, Italy; General Surgical Oncology Department, Istituto di Ricovero e Cura a Carattere Scientifico Centro di Riferimento Oncologico di Aviano (Istituto Nazionale Tumori), Aviano, Italy.
| | - Vittorio Bresadola
- Department of Medicine, General Surgery Department and Simulation Center, Academic Hospital of Udine, University of Udine, Udine, Italy
| |
Collapse
|
10
|
Mohizin A, Imran JH, Lee KS, Kim JK. Dynamic interaction of injected liquid jet with skin layer interfaces revealed by microsecond imaging of optically cleared ex vivo skin tissue model. J Biol Eng 2023; 17:15. [PMID: 36849998 PMCID: PMC9969392 DOI: 10.1186/s13036-023-00335-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/21/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Needle-free jet injection (NFJI) systems enable a controlled and targeted delivery of drugs into skin tissue. However, a scarce understanding of their underlying mechanisms has been a major deterrent to the development of an efficient system. Primarily, the lack of a suitable visualization technique that could capture the dynamics of the injected fluid-tissue interaction with a microsecond range temporal resolution has emerged as a main limitation. A conventional needle-free injection system may inject the fluids within a few milliseconds and may need a temporal resolution in the microsecond range for obtaining the required images. However, the presently available imaging techniques for skin tissue visualization fail to achieve these required spatial and temporal resolutions. Previous studies on injected fluid-tissue interaction dynamics were conducted using in vitro media with a stiffness similar to that of skin tissue. However, these media are poor substitutes for real skin tissue, and the need for an imaging technique having ex vivo or in vivo imaging capability has been echoed in the previous reports. METHODS A near-infrared imaging technique that utilizes the optical absorption and fluorescence emission of indocyanine green dye, coupled with a tissue clearing technique, was developed for visualizing a NFJI in an ex vivo porcine skin tissue. RESULTS The optimal imaging conditions obtained by considering the optical properties of the developed system and mechanical properties of the cleared ex vivo samples are presented. Crucial information on the dynamic interaction of the injected liquid jet with the ex vivo skin tissue layers and their interfaces could be obtained. CONCLUSIONS The reported technique can be instrumental for understanding the injection mechanism and for the development of an efficient transdermal NFJI system as well.
Collapse
Affiliation(s)
- Abdul Mohizin
- School of Mechanical Engineering, Kookmin University, 77 Jeongneung-Ro, Seongbuk-Gu, Seoul, 02707, Republic of Korea
| | - Jakir Hossain Imran
- Department of Mechanical Engineering, Graduate School, Kookmin University, Seoul, 02707, Republic of Korea
| | - Kee Sung Lee
- School of Mechanical Engineering, Kookmin University, 77 Jeongneung-Ro, Seongbuk-Gu, Seoul, 02707, Republic of Korea
| | - Jung Kyung Kim
- School of Mechanical Engineering, Kookmin University, 77 Jeongneung-Ro, Seongbuk-Gu, Seoul, 02707, Republic of Korea.
| |
Collapse
|
11
|
Li K, Wu S, Dong G, Li Y, Wang W, Dong G, Hong Z, Li M, Sheng C. Environmentally sensitive fluorescent probes with improved properties for detecting and imaging PDEδ in live cells and tumor slices. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
12
|
de Vries E, Alic L, Schols RM, Emanuel KS, Wieringa FP, Bouvy ND, Tuijthof GJM. Near-Infrared Spectral Similarity between Ex Vivo Porcine and In Vivo Human Tissue. Life (Basel) 2023; 13:life13020357. [PMID: 36836713 PMCID: PMC9959888 DOI: 10.3390/life13020357] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND In vivo diffuse reflectance spectroscopy provides additional contrast in discriminating nerves embedded in adipose tissue during surgery. However, large datasets are required to achieve clinically acceptable classification levels. This study assesses the spectral similarity between ex vivo porcine and in vivo human spectral data of nerve and adipose tissue, as porcine tissue could contribute to generate large datasets. METHODS Porcine diffuse reflectance spectra were measured at 124 nerve and 151 adipose locations. A previously recorded dataset of 32 in vivo human nerve and 23 adipose tissue locations was used for comparison. In total, 36 features were extracted from the raw porcine to generate binary logistic regression models for all combinations of two, three, four and five features. Feature selection was performed by assessing similar means between normalized features of nerve and of adipose tissue (Kruskal-Wallis test, p < 0.05) and for models performing best on the porcine cross validation set. The human test set was used to assess classification performance. RESULTS The binary logistic regression models with selected features showed an accuracy of 60% on the test set. CONCLUSIONS Spectral similarity between ex vivo porcine and in vivo human adipose and nerve tissue was present, but further research is required.
Collapse
Affiliation(s)
- Eva de Vries
- Research Engineering, Faculty of Health, Medicine, Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Lejla Alic
- Magnetic Detection and Imaging Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
| | - Rutger M. Schols
- Department of Plastic, Reconstructive and Hand Surgery, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Kaj S. Emanuel
- Department of Orthopaedic Surgery, Faculty of Health, Medicine, Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Orthopedic Surgery and Sports Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, 1105 AZ Amsterdam, The Netherlands
| | | | - Nicole D. Bouvy
- Department of Surgery, School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Gabriëlle J. M. Tuijthof
- Research Engineering, Faculty of Health, Medicine, Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Biomechanical Engineering, Faculty of Engineering Technologies, University of Twente, 7522 NB Enschede, The Netherlands
- Correspondence: ; Tel.: +31-639265645
| |
Collapse
|
13
|
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
|
14
|
Demarchi MS, Karenovics W, Bédat B, Triponez F. Near-infrared fluorescent imaging techniques for the detection and preservation of parathyroid glands during endocrine surgery. Innov Surg Sci 2022; 7:87-98. [PMID: 36561508 PMCID: PMC9742281 DOI: 10.1515/iss-2021-0001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 04/14/2021] [Indexed: 12/25/2022] Open
Abstract
Objectives In over 30% of all thyroid surgeries, complications arise from transient and definitive hypoparathyroidism, underscoring the need for real-time identification and preservation of parathyroid glands (PGs). Here, we evaluate the promising intraoperative optical technologies available for the identification, preservation, and functional assessment of PGs to enhance endocrine surgery. Methods We performed a review of the literature to identify published studies on fluorescence imaging in thyroid and parathyroid surgery. Results Fluorescence imaging is a well-demonstrated approach for both in vivo and in vitro localization of specific cells or tissues, and is gaining popularity as a technique to detect PGs during endocrine surgery. Autofluorescence (AF) imaging and indocyanine green (ICG) angiography are two emerging optical techniques to improve outcomes in thyroid and parathyroid surgeries. Near-infrared-guided technology has significantly contributed to the localization of PGs, through the detection of glandular AF. Perfusion through the PGs can be visualized with ICG, which can also reveal the blood supply after dissection. Conclusions Near infrared AF and ICG angiography, providing a valuable spatial and anatomical information, can decrease the incidence of complications in thyroid surgery.
Collapse
Affiliation(s)
- Marco Stefano Demarchi
- Department of Thoracic and Endocrine Surgery, Faculty of Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Wolfram Karenovics
- Department of Thoracic and Endocrine Surgery, Faculty of Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Benoît Bédat
- Department of Thoracic and Endocrine Surgery, Faculty of Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Frédéric Triponez
- Department of Thoracic and Endocrine Surgery, Faculty of Medicine, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
15
|
O'Brien CM, Bishop KW, Zhang H, Xu X, Shmuylovich L, Conley E, Nwosu K, Duncan K, Mondal SB, Sudlow G, Achilefu S. Quantitative tumor depth determination using dual wavelength excitation fluorescence. BIOMEDICAL OPTICS EXPRESS 2022; 13:5628-5642. [PMID: 36733737 PMCID: PMC9872884 DOI: 10.1364/boe.468059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 06/07/2023]
Abstract
Quantifying solid tumor margins with fluorescence-guided surgery approaches is a challenge, particularly when using near infrared (NIR) wavelengths due to increased penetration depths. An NIR dual wavelength excitation fluorescence (DWEF) approach was developed that capitalizes on the wavelength-dependent attenuation of light in tissue to determine fluorophore depth. A portable dual wavelength excitation fluorescence imaging system was built and tested in parallel with an NIR tumor-targeting fluorophore in tissue mimicking phantoms, chicken tissue, and in vivo mouse models of breast cancer. The system showed high accuracy in all experiments. The low cost and simplicity of this approach make it ideal for clinical use.
Collapse
Affiliation(s)
- Christine M O'Brien
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive St. Louis, MO 63130, USA
- These authors contributed equally to this work
| | - Kevin W Bishop
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
| | - Haini Zhang
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive St. Louis, MO 63130, USA
| | - Xiao Xu
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
| | - Leo Shmuylovich
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, 4960 Children's Place, St. Louis, MO 63110, USA
| | - Elizabeth Conley
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
| | - Karen Nwosu
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
| | - Kathleen Duncan
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
| | - Suman B Mondal
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
| | - Gail Sudlow
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive St. Louis, MO 63130, USA
- Department of Medicine, Washington University School of Medicine, 4960 Children's Place, St. Louis, MO 63110, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
- These authors contributed equally to this work
| |
Collapse
|
16
|
Indocyanine Green Endoscopy for Pituitary Adenomas with Parasellar Extension: Results from a Preliminary Case Series. World Neurosurg 2022; 166:e692-e702. [PMID: 35917924 DOI: 10.1016/j.wneu.2022.07.081] [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: 03/05/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Advances in visualization tools have brought new confidence, including endoscope-integrated indocyanine (E-ICG), which makes pituitary and skull-base surgery safer and more effective. We report here our preliminary experience with the use of E-ICG to 1) visualize the cavernous segment of the internal carotid artery (ICA); and 2) functionally and anatomically preserve the pituitary gland. METHODS A dedicated ICG-integrated endoscope was used in 15 patients with parasellar pituitary adenomas. Indocyanine was administered at 2 different time points during surgery: an early bolus of 12.5 mg at the sphenoid sinus opening to expose the position of the parasellar segment of the ICAs and to identify the position of the normal pituitary gland so that it could be preserved during tumor removal. Subsequently, a second late bolus of 12 mg of ICG was injected to obtain a real-time "wire angiographic" visualization of the flow of the ICAs. RESULTS Gross total resection was achieved in 12 cases (80%), whereas subtotal resection was performed in the other 3 cases (20%). The pituitary gland was clearly discernable in 11 cases (91.6%). None of the patients manifested new endocrinologic deficits or major vascular complications. CONCLUSIONS E-ICG is a safe and essential aid for pituitary adenomas invading the cavernous sinus. Its performance as a pituitary marker and real-time video angiography showed promising results in terms of extent of resection, endocrinologic outcomes, and prevention of intraoperative complications.
Collapse
|
17
|
Boekestijn I, van Oosterom MN, Dell'Oglio P, van Velden FHP, Pool M, Maurer T, Rietbergen DDD, Buckle T, van Leeuwen FWB. The current status and future prospects for molecular imaging-guided precision surgery. Cancer Imaging 2022; 22:48. [PMID: 36068619 PMCID: PMC9446692 DOI: 10.1186/s40644-022-00482-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 08/21/2022] [Indexed: 01/19/2023] Open
Abstract
Molecular imaging technologies are increasingly used to diagnose, monitor, and guide treatment of i.e., cancer. In this review, the current status and future prospects of the use of molecular imaging as an instrument to help realize precision surgery is addressed with focus on the main components that form the conceptual basis of intraoperative molecular imaging. Paramount for successful interventions is the relevance and accessibility of surgical targets. In addition, selection of the correct combination of imaging agents and modalities is critical to visualize both microscopic and bulk disease sites with high affinity and specificity. In this context developments within engineering/imaging physics continue to drive the growth of image-guided surgery. Particularly important herein is enhancement of sensitivity through improved contrast and spatial resolution, features that are critical if sites of cancer involvement are not to be overlooked during surgery. By facilitating the connection between surgical planning and surgical execution, digital surgery technologies such as computer-aided visualization nicely complement these technologies. The complexity of image guidance, combined with the plurality of technologies that are becoming available, also drives the need for evaluation mechanisms that can objectively score the impact that technologies exert on the performance of healthcare professionals and outcome improvement for patients.
Collapse
Affiliation(s)
- Imke Boekestijn
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Paolo Dell'Oglio
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Urology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Floris H P van Velden
- Medical Physics, Department of Radiology , Leiden University Medical Center, Leiden, the Netherlands
| | - Martin Pool
- Department of Clinical Farmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Centre Hamburg, Hamburg, Germany
| | - Daphne D D Rietbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
18
|
Martin E, Hom M, Mani L, Rosenthal EL. Current and Future Applications of Fluorescence-Guided Surgery in Head and Neck Cancer. Surg Oncol Clin N Am 2022; 31:695-706. [DOI: 10.1016/j.soc.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
19
|
Xing J, Gong Q, Zou R, Yao J, Xiang L, Wu A. GSH responsive traditional clinical drugs probe for cancer cell fluorescence imaging and therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
20
|
Yokomizo S, Henary M, Buabeng ER, Fukuda T, Monaco H, Baek Y, Manganiello S, Wang H, Kubota J, Ulumben AD, Lv X, Wang C, Inoue K, Fukushi M, Kang H, Bao K, Kashiwagi S, Choi HS. Topical pH Sensing NIR Fluorophores for Intraoperative Imaging and Surgery of Disseminated Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201416. [PMID: 35567348 PMCID: PMC9286000 DOI: 10.1002/advs.202201416] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Indexed: 05/05/2023]
Abstract
Fluorescence-guided surgery (FGS) aids surgeons with real-time visualization of small cancer foci and borders, which improves surgical and prognostic efficacy of cancer. Despite the steady advances in imaging devices, there is a scarcity of fluorophores available to achieve optimal FGS. Here, 1) a pH-sensitive near-infrared fluorophore that exhibits rapid signal changes in acidic tumor microenvironments (TME) caused by the attenuation of intramolecular quenching, 2) the inherent targeting for cancer based on chemical structure (structure inherent targeting, SIT), and 3) mitochondrial and lysosomal retention are reported. After topical application of PH08 on peritoneal tumor regions in ovarian cancer-bearing mice, a rapid fluorescence increase (< 10 min), and extended preservation of signals (> 4 h post-topical application) are observed, which together allow for the visualization of submillimeter tumors with a high tumor-to-background ratio (TBR > 5.0). In addition, PH08 is preferentially transported to cancer cells via organic anion transporter peptides (OATPs) and colocalizes in the mitochondria and lysosomes due to the positive charges, enabling a long retention time during FGS. PH08 not only has a significant impact on surgical and diagnostic applications but also provides an effective and scalable strategy to design therapeutic agents for a wide array of cancers.
Collapse
Affiliation(s)
- Shinya Yokomizo
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
- Department of Radiological SciencesTokyo Metropolitan University7‐2‐10 Higashi‐OguArakawaTokyo116–8551Japan
| | - Maged Henary
- Department of Chemistry and Center for Diagnostics and TherapeuticsGeorgia State University100 Piedmont Avenue SEAtlantaGA30303USA
| | - Emmanuel R. Buabeng
- Department of Chemistry and Center for Diagnostics and TherapeuticsGeorgia State University100 Piedmont Avenue SEAtlantaGA30303USA
| | - Takeshi Fukuda
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
- Department of Obstetrics and GynecologyOsaka City University Graduate School of Medicine1‐4‐3, AsahimachiAbeno‐kuOsaka545–8585Japan
| | - Hailey Monaco
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Yoonji Baek
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Sophia Manganiello
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Haoran Wang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Jo Kubota
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Amy Daniel Ulumben
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Xiangmin Lv
- Vincent Center for Reproductive BiologyVincent Department of Obstetrics and GynecologyMassachusetts General HospitalBostonMA02114USA
| | - Cheng Wang
- Vincent Center for Reproductive BiologyVincent Department of Obstetrics and GynecologyMassachusetts General HospitalBostonMA02114USA
| | - Kazumasa Inoue
- Department of Radiological SciencesTokyo Metropolitan University7‐2‐10 Higashi‐OguArakawaTokyo116–8551Japan
| | - Masahiro Fukushi
- Department of Radiological SciencesTokyo Metropolitan University7‐2‐10 Higashi‐OguArakawaTokyo116–8551Japan
| | - Homan Kang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Kai Bao
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Hak Soo Choi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| |
Collapse
|
21
|
Lauwerends LJ, Abbasi H, Bakker Schut TC, Van Driel PBAA, Hardillo JAU, Santos IP, Barroso EM, Koljenović S, Vahrmeijer AL, Baatenburg de Jong RJ, Puppels GJ, Keereweer S. The complementary value of intraoperative fluorescence imaging and Raman spectroscopy for cancer surgery: combining the incompatibles. Eur J Nucl Med Mol Imaging 2022; 49:2364-2376. [PMID: 35102436 PMCID: PMC9165240 DOI: 10.1007/s00259-022-05705-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/23/2022] [Indexed: 01/09/2023]
Abstract
A clear margin is an important prognostic factor for most solid tumours treated by surgery. Intraoperative fluorescence imaging using exogenous tumour-specific fluorescent agents has shown particular benefit in improving complete resection of tumour tissue. However, signal processing for fluorescence imaging is complex, and fluorescence signal intensity does not always perfectly correlate with tumour location. Raman spectroscopy has the capacity to accurately differentiate between malignant and healthy tissue based on their molecular composition. In Raman spectroscopy, specificity is uniquely high, but signal intensity is weak and Raman measurements are mainly performed in a point-wise manner on microscopic tissue volumes, making whole-field assessment temporally unfeasible. In this review, we describe the state-of-the-art of both optical techniques, paying special attention to the combined intraoperative application of fluorescence imaging and Raman spectroscopy in current clinical research. We demonstrate how these techniques are complementary and address the technical challenges that have traditionally led them to be considered mutually exclusive for clinical implementation. Finally, we present a novel strategy that exploits the optimal characteristics of both modalities to facilitate resection with clear surgical margins.
Collapse
Affiliation(s)
- L J Lauwerends
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - H Abbasi
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
- Center for Optical Diagnostics and Therapy, Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - T C Bakker Schut
- Center for Optical Diagnostics and Therapy, Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - P B A A Van Driel
- Department of Orthopedic Surgery, Isala Hospital, Zwolle, Netherlands
| | - J A U Hardillo
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - I P Santos
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | | | - S Koljenović
- Department of Pathology, Antwerp University Hospital/Antwerp University, Antwerp, Belgium
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - R J Baatenburg de Jong
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - G J Puppels
- Center for Optical Diagnostics and Therapy, Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - S Keereweer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands.
| |
Collapse
|
22
|
Vergeer RA, Theunissen REP, van Elk T, Schmidt I, Postma MR, Tamasi K, van Dijk JMC, Kuijlen JMA. Fluorescence-guided detection of pituitary neuroendocrine tumor (PitNET) tissue during endoscopic transsphenoidal surgery available agents, their potential, and technical aspects. Rev Endocr Metab Disord 2022; 23:647-657. [PMID: 35344185 PMCID: PMC9156450 DOI: 10.1007/s11154-022-09718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 10/29/2022]
Abstract
Differentiation of pituitary neuroendocrine tumor (PitNET) tissue from surrounding normal tissue during surgery is challenging. A number of fluorescent agents is available for visualization of tissue discrepancy, with the potential of improving total tumor resection. This review evaluates the availability, clinical and technical applicability of the various fluorescent agents within the field of pituitary surgery. According to PRISMA guidelines, a systematic review was performed to identify reports describing results of in vivo application of fluorescent agents. In this review, 15 publications were included. Sodium Fluorescein (FNa) was considered in two studies. The first study reported noticeable fluorescence in adenoma tissue, the second demonstrated the strongest fluorescence in non-functioning pituitary adenomas. 5-Aminolevulinic acid (5-ALA) was investigated in three studies. One study compared laser-based optical biopsy system (OBS) with photo-diagnostic filter (PD) and found that the OBS was able to detect all microadenomas, even when MRI was negative. The second study retrospectively analyzed twelve pituitary adenomas and found only one positive for fluorescence. The third investigated fifteen pituitary adenomas of which one displayed vague fluorescence. Indocyanine green (ICG) was researched in four studies with variable results. Second-Window ICG yielded no significant difference between functioning and non-functioning adenomas in one study, while a second study displayed 4 times higher fluorescence in tumor tissue than in normal tissue. In three studies, OTL38 showed potential in non-functioning pituitary adenomas. At present, evidence for fluorescent agents to benefit total resection of PitNETs is lacking. OTL38 can potentially serve as a selective fluorescent agent in non-functioning pituitary adenomas in the near future.
Collapse
Affiliation(s)
- Rob A Vergeer
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Robin E P Theunissen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Theodora van Elk
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mark R Postma
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Katalin Tamasi
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J Marc C van Dijk
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jos M A Kuijlen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Wilson BC, Eu D. Optical Spectroscopy and Imaging in Surgical Management of Cancer Patients. TRANSLATIONAL BIOPHOTONICS 2022. [DOI: 10.1002/tbio.202100009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Brian C. Wilson
- Princess Margaret Cancer Centre/University Health Network 101 College Street Toronto Ontario Canada
- Department of Medical Biophysics, Faculty of Medicine University of Toronto Canada
| | - Donovan Eu
- Department of Otolaryngology‐Head and Neck Surgery‐Surgical Oncology, Princess Margaret Cancer Centre/University Health Network University of Toronto Canada
- Department of Otolaryngology‐Head and Neck Surgery National University Hospital System Singapore
| |
Collapse
|
24
|
Najafiaghdam H, Rabbani R, Gharia A, Papageorgiou EP, Anwar M. 3D Reconstruction of cellular images from microfabricated imagers using fully-adaptive deep neural networks. Sci Rep 2022; 12:7229. [PMID: 35508477 PMCID: PMC9068918 DOI: 10.1038/s41598-022-10886-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/24/2022] [Indexed: 11/09/2022] Open
Abstract
Millimeter-scale multi-cellular level imagers enable various applications, ranging from intraoperative surgical navigation to implantable sensors. However, the tradeoffs for miniaturization compromise resolution, making extracting 3D cell locations challenging—critical for tumor margin assessment and therapy monitoring. This work presents three machine-learning-based modules that extract spatial information from single image acquisitions using custom-made millimeter-scale imagers. The neural networks were trained on synthetically-generated (using Perlin noise) cell images. The first network is a convolutional neural network estimating the depth of a single layer of cells, the second is a deblurring module correcting for the point spread function (PSF). The final module extracts spatial information from a single image acquisition of a 3D specimen and reconstructs cross-sections, by providing a layered “map” of cell locations. The maximum depth error of the first module is 100 µm, with 87% test accuracy. The second module’s PSF correction achieves a least-square-error of only 4%. The third module generates a binary “cell” or “no cell” per-pixel labeling with an accuracy ranging from 89% to 85%. This work demonstrates the synergy between ultra-small silicon-based imagers that enable in vivo imaging but face a trade-off in spatial resolution, and the processing power of neural networks to achieve enhancements beyond conventional linear optimization techniques.
Collapse
Affiliation(s)
- Hossein Najafiaghdam
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, 94720, USA.
| | - Rozhan Rabbani
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, 94720, USA
| | - Asmaysinh Gharia
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, 94720, USA
| | - Efthymios P Papageorgiou
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, 94720, USA
| | - Mekhail Anwar
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, 94720, USA.,Department of Radiation Oncology, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
25
|
Voskuil FJ, Vonk J, van der Vegt B, Kruijff S, Ntziachristos V, van der Zaag PJ, Witjes MJH, van Dam GM. Intraoperative imaging in pathology-assisted surgery. Nat Biomed Eng 2022; 6:503-514. [PMID: 34750537 DOI: 10.1038/s41551-021-00808-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
The pathological assessment of surgical specimens during surgery can reduce the incidence of positive resection margins, which otherwise can result in additional surgeries or aggressive therapeutic regimens. To improve patient outcomes, intraoperative spectroscopic, fluorescence-based, structural, optoacoustic and radiological imaging techniques are being tested on freshly excised tissue. The specific clinical setting and tumour type largely determine whether endogenous or exogenous contrast is to be detected and whether the tumour specificity of the detected biomarker, image resolution, image-acquisition times or penetration depth are to be prioritized. In this Perspective, we describe current clinical standards for intraoperative tissue analysis and discuss how intraoperative imaging is being implemented. We also discuss potential implementations of intraoperative pathology-assisted surgery for clinical decision-making.
Collapse
Affiliation(s)
- Floris J Voskuil
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jasper Vonk
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Schelto Kruijff
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vasilis Ntziachristos
- Chair for Biological Imaging, Center for Translational Cancer Research, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Pieter J van der Zaag
- Phillips Research Laboratories, Eindhoven, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Molecular Biophysics, Zernike Institute, University of Groningen, Groningen, The Netherlands
| | - Max J H Witjes
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gooitzen M van Dam
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. .,AxelaRx/TRACER BV, Groningen, The Netherlands.
| |
Collapse
|
26
|
Fukuda T, Yokomizo S, Casa S, Monaco H, Manganiello S, Wang H, Lv X, Ulumben AD, Yang C, Kang MW, Inoue K, Fukushi M, Sumi T, Wang C, Kang H, Bao K, Henary M, Kashiwagi S, Soo Choi H. Fast and Durable Intraoperative Near-infrared Imaging of Ovarian Cancer Using Ultrabright Squaraine Fluorophores. Angew Chem Int Ed Engl 2022; 61:e202117330. [PMID: 35150468 PMCID: PMC9007913 DOI: 10.1002/anie.202117330] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Indexed: 12/19/2022]
Abstract
The residual tumor after surgery is the most significant prognostic factor of patients with epithelial ovarian cancer. Near-infrared (NIR) fluorescence-guided surgery is actively utilized for tumor localization and complete resection during surgery. However, currently available contrast-enhancing agents display low on-target binding, unfavorable pharmacokinetics, and toxicity, thus not ideal for clinical use. Here we report ultrabright and stable squaraine fluorophores with optimal pharmacokinetics by introducing an asymmetric molecular conformation and surface charges for rapid transporter-mediated cellular uptake. Among the tested, OCTL14 shows low serum binding and rapid distribution into cancer tissue via organic cation transporters (OCTs). Additionally, the charged squaraine fluorophores are retained in lysosomes, providing durable intraoperative imaging in a preclinical murine model of ovarian cancer up to 24 h post-injection. OCTL14 represents a significant departure from the current bioconjugation approach of using a non-targeted fluorophore and would provide surgeons with an indispensable tool to achieve optimal resection.
Collapse
Affiliation(s)
- Takeshi Fukuda
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, 1-4-3, Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shinya Yokomizo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo, 116-8551, Japan
| | - Stefanie Casa
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Hailey Monaco
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Sophia Manganiello
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Haoran Wang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Xiangmin Lv
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amy Daniel Ulumben
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Chengeng Yang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Min-Woong Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Chungnam National University, Daejeon, 301-721, South Korea
| | - Kazumasa Inoue
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo, 116-8551, Japan
| | - Masahiro Fukushi
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo, 116-8551, Japan
| | - Toshiyuki Sumi
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, 1-4-3, Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kai Bao
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Maged Henary
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
- Center for Diagnostics and Therapeutics, 145 Piedmont Avenue S.E., Atlanta, GA 30303, USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
27
|
Ren W, Ji B, Guan Y, Cao L, Ni R. Recent Technical Advances in Accelerating the Clinical Translation of Small Animal Brain Imaging: Hybrid Imaging, Deep Learning, and Transcriptomics. Front Med (Lausanne) 2022; 9:771982. [PMID: 35402436 PMCID: PMC8987112 DOI: 10.3389/fmed.2022.771982] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/16/2022] [Indexed: 12/26/2022] Open
Abstract
Small animal models play a fundamental role in brain research by deepening the understanding of the physiological functions and mechanisms underlying brain disorders and are thus essential in the development of therapeutic and diagnostic imaging tracers targeting the central nervous system. Advances in structural, functional, and molecular imaging using MRI, PET, fluorescence imaging, and optoacoustic imaging have enabled the interrogation of the rodent brain across a large temporal and spatial resolution scale in a non-invasively manner. However, there are still several major gaps in translating from preclinical brain imaging to the clinical setting. The hindering factors include the following: (1) intrinsic differences between biological species regarding brain size, cell type, protein expression level, and metabolism level and (2) imaging technical barriers regarding the interpretation of image contrast and limited spatiotemporal resolution. To mitigate these factors, single-cell transcriptomics and measures to identify the cellular source of PET tracers have been developed. Meanwhile, hybrid imaging techniques that provide highly complementary anatomical and molecular information are emerging. Furthermore, deep learning-based image analysis has been developed to enhance the quantification and optimization of the imaging protocol. In this mini-review, we summarize the recent developments in small animal neuroimaging toward improved translational power, with a focus on technical improvement including hybrid imaging, data processing, transcriptomics, awake animal imaging, and on-chip pharmacokinetics. We also discuss outstanding challenges in standardization and considerations toward increasing translational power and propose future outlooks.
Collapse
Affiliation(s)
- Wuwei Ren
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Engineering Research Center of Energy Efficient and Custom AI IC, Shanghai, China
| | - Bin Ji
- Department of Radiopharmacy and Molecular Imaging, School of Pharmacy, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Cao
- Shanghai Changes Tech, Ltd., Shanghai, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, ETH Zürich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Fukuda T, Yokomizo S, Casa S, Monaco H, Manganiello S, Wang H, Lv X, Ulumben AD, Yang C, Kang MW, Inoue K, Fukushi M, Sumi T, Wang C, Kang H, Bao K, Henary M, Kashiwagi S, Choi HS. Fast and Durable Intraoperative Near‐infrared Imaging of Ovarian Cancer Using Ultrabright Squaraine Fluorophores. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202117330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | | | | | - Hailey Monaco
- Massachusetts General Hospital radiology UNITED STATES
| | | | - Haoran Wang
- Massachusetts General Hospital radiology UNITED STATES
| | - Xiangmin Lv
- Massachusetts General Hospital Obstetrics and Gynecology UNITED STATES
| | | | - Chengeng Yang
- Massachusetts General Hospital radiology UNITED STATES
| | | | - Kazumasa Inoue
- Tokyo Metropolitan University - Arakawa Campus: Tokyo Toritsu Daigaku - Arakawa Campus Radiation Science JAPAN
| | - Masahiro Fukushi
- Tokyo Metropolitan University - Arakawa Campus: Tokyo Toritsu Daigaku - Arakawa Campus Radiation Science JAPAN
| | - Toshiyuki Sumi
- Osaka City University: Osaka Shiritsu Daigaku Obstetrics and Gynecology JAPAN
| | - Cheng Wang
- Massachusetts General Hospital Obstetrics and Gynecology UNITED STATES
| | - Homan Kang
- Massachusetts General Hospital radiology UNITED STATES
| | - Kai Bao
- Massachusetts General Hospital radiology UNITED STATES
| | - Maged Henary
- Georgia State University Chemistry UNITED STATES
| | - Satoshi Kashiwagi
- Massachusetts General Hospital Radiology 149 13th Street 02129 Charlestown UNITED STATES
| | - Hak Soo Choi
- Massachusetts General Hospital Radiology 149 13th Street 02129 Boston UNITED STATES
| |
Collapse
|
29
|
Mansi M, Howley R, Chen B. Methods to Measure the Inhibition of ABCG2 Transporter and Ferrochelatase Activity to Enhance Aminolevulinic Acid-Protoporphyrin IX Fluorescence-Guided Tumor Detection and Resection. Methods Mol Biol 2022; 2394:823-835. [PMID: 35094360 DOI: 10.1007/978-1-0716-1811-0_43] [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] [Indexed: 01/25/2023]
Abstract
Aminolevulinic acid (ALA) has been clinically used as an intraoperative fluorescence probe for protoporphyrin IX (PpIX) fluorescence-guided tumor resection and a PDT agent for cancer treatment. Although tumor tissues often show increased ALA-PpIX fluorescence compared with normal tissues, which enables the use of ALA for tumor imaging and targeting, weak tumor PpIX fluorescence as well as the heterogeneity in tumor fluorescence severely limits its clinical application. Intracellular PpIX in tumor cells is reduced by two major mechanisms, efflux by ATP-binding cassette (ABC) transporters such as ABCG2 and bioconversion to form heme by ferrochelatase (FECH) in the heme biosynthesis pathway. Targeting these two predominant PpIX-reducing mechanisms for the enhancement of ALA-PpIX have yielded a plethora of promising results and stimulated the clinical exploration of these enhancement strategies. Here we describe our methods of evaluating chemicals for the inhibition of ABCG2 transporter and FECH activity. Our goal is to further encourage research and development of novel ABCG2 and FECH inhibitors and promote a rational use of these inhibitors to optimize ALA-based tumor detection and treatment.
Collapse
Affiliation(s)
- Matthew Mansi
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA
| | - Richard Howley
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA
| | - Bin Chen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA, USA. .,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
30
|
Mickelson MA. Updated Concepts in Oncologic Surgery: Apocrine Gland Anal Sac Adenocarcinoma and Mast Cell Tumors. Vet Clin North Am Small Anim Pract 2022; 52:549-580. [DOI: 10.1016/j.cvsm.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
Merkes JM, Hasenbach A, Kiessling F, Hermann S, Banala S. Sensing Reactive Oxygen Species with Photoacoustic Imaging Using Conjugation-Extended BODIPYs. ACS Sens 2021; 6:4379-4388. [PMID: 34898171 DOI: 10.1021/acssensors.1c01674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Short-lived reactive intermediates such as reactive oxygen species (ROS) regulate many physiological processes, but overproduction can also lead to severe tissue dysfunction. Thus, there is a high demand for noninvasive detection of reactive molecules, which, however, is challenging. Herein, we report photoacoustic detection of ROS using conjugated BODIPY probes (ROS-BODIPYs). The ROS reaction with conjugated BODIPYs induced a redshift in absorption by ∼100 nm into the near infrared (from ∼700 to ∼800 nm), quenched fluorescence, and generated strong photoacoustic (PA) signals. Thus, the ROS-activated and ROS-nonactivated states of ROS-BODIPYs can be detected in vivo by PA and fluorescence imaging. Interestingly, ROS activation is reversible, in the presence of excess reducing agents, e.g., citric acid, converted back to its original state, suggesting that ROS-BODIPYs can be useful for the detection of over production of ROS but not physiological amounts. This makes the imaging independent of accumulation of the activated probe with the physiological ROS amounts and thus strongly improves applicability and highlights the translational potential of ROS-BODIPYs for detecting overexpression of ROS in vivo by optical and photoacoustic imaging methods.
Collapse
Affiliation(s)
- Jean Michél Merkes
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074 Aachen, Germany
- Institute for Experimental Molecular Imaging University Clinic, RWTH Aachen University, Forckenbeckstra. 55, 52074 Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Str. 2, 28359 Bremen, Germany
| | - Alexa Hasenbach
- European Institute for Molecular Imaging, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging University Clinic, RWTH Aachen University, Forckenbeckstra. 55, 52074 Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Str. 2, 28359 Bremen, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging, University of Münster, Waldeyerstr. 15, 48149 Münster, Germany
| | - Srinivas Banala
- Institute of Organic Chemistry, RWTH Aachen University, Landoltweg 1, 52074 Aachen, Germany
- Institute for Experimental Molecular Imaging University Clinic, RWTH Aachen University, Forckenbeckstra. 55, 52074 Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Max-von-Laue-Str. 2, 28359 Bremen, Germany
| |
Collapse
|
32
|
Privitera L, Paraboschi I, Cross K, Giuliani S. Above and Beyond Robotic Surgery and 3D Modelling in Paediatric Cancer Surgery. Front Pediatr 2021; 9:777840. [PMID: 34988038 PMCID: PMC8721224 DOI: 10.3389/fped.2021.777840] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Although the survival rates for children's cancers have more than doubled in the last few decades, the surgical practise has not significantly changed. Among the most recent innovations introduced in the clinic, robotic surgery and augmented reality are two of the most promising, even if they are not widespread. The increased flexibility of the motion, the magnification of the surgical field and the tremor reduction provided by robotic surgery have been beneficial to perform complex oncological procedures in children. Besides, augmented reality has been proven helpful in planning for tumour removal, facilitating early discrimination between cancer and healthy organs. Nowadays, research in the field of surgical oncology is moving fast, and new technologies and innovations wich will help to shape a new way to perform cancer surgery. Paediatric surgeons need to be ready to adopt these novel devices and intraoperative techniques to allow more radical tumour resections with fewer complications. This review aims to present the mechanism of action and indications of several novel technologies such as optical imaging surgery, high definition cameras, and intraoperative loco-regional treatments. We hope this will enhance early adoption and more research on how to employ technology for the benefit of children.
Collapse
Affiliation(s)
- Laura Privitera
- Wellcome/Engineering and Physical Sciences Research Council Centre for Interventional & Surgical Sciences, University College London, London, United Kingdom
- Developmental Biology and Cancer Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Irene Paraboschi
- Wellcome/Engineering and Physical Sciences Research Council Centre for Interventional & Surgical Sciences, University College London, London, United Kingdom
- Developmental Biology and Cancer Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Kate Cross
- Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Stefano Giuliani
- Wellcome/Engineering and Physical Sciences Research Council Centre for Interventional & Surgical Sciences, University College London, London, United Kingdom
- Developmental Biology and Cancer Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
33
|
Borlan R, Focsan M, Perde-Schrepler M, Soritau O, Campu A, Gaina L, Pall E, Pop B, Baldasici O, Gherman C, Stoia D, Maniu D, Astilean S. Antibody-functionalized theranostic protein nanoparticles for the synergistic deep red fluorescence imaging and multimodal therapy of ovarian cancer. Biomater Sci 2021; 9:6183-6202. [PMID: 34346411 DOI: 10.1039/d1bm01002f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Among women, ovarian cancer is the fifth most frequent type of cancer, and despite benefiting from current standard treatment plans, 90% of patients relapse in the subsequent 18 months and, eventually, perish. As a result, via embracing nanotechnological advancements in the field of medical science, researchers working in the areas of cancer therapy and imaging are looking for the next breakthrough treatment strategy to ensure lower cancer recurrence rates and improved outcomes for patients. Herein, we design a novel phototheranostic agent with optical features in the biological window of the electromagnetic spectrum via encapsulating a newly synthesized phthalocyanine dye within biocompatible protein nanoparticles, allowing the targeted fluorescence imaging and synergistic dual therapy of ovarian cancer. The nanosized agent displays great biocompatibility and enhanced aqueous biostability and photothermal activity, as well as high reactive-oxygen-species generation efficiency. To achieve the active targeting of the desired malignant tissue and suppress the rapid clearance of the photosensitive agent from the peritoneal cavity, the nanoparticles are biofunctionalized with an anti-folate receptor antibody. A2780 ovarian cancer cells are employed to confirm the improved targeting capabilities and the in vitro cytotoxic efficiency of the theranostic nanoparticles after exposure to a 660 nm LED lamp; upon measurement via MTT and flow cytometry assays, a significant 95% decrease in the total number of viable cells is seen. Additionally, the therapeutic performance of our newly designed nanoparticles was evaluated in vivo, via real-time thermal monitoring and histopathological assays, upon the irradiation of tumour-bearing mice with a 660 nm LED lamp (0.05 W cm-2). Foremost, separately from steady-state fluorescence imaging, we found that, via utilizing FLIM investigations, the differences in fluorescence lifetimes of antibody biofunctionalized and non-functionalized nanoparticles can be correlated to different intracellular localization and internalization pathways of the fluorescent agent, which is relevant for the development of a cutting-edge method for the detection of cancer cells that overexpress folate receptors at their surfaces.
Collapse
Affiliation(s)
- Raluca Borlan
- Biomolecular Physics Department, Faculty of Physics, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania. and Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania.
| | - Monica Focsan
- Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania.
| | - Maria Perde-Schrepler
- Department of Radiobiology and Tumor Biology, Oncology Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Cluj, Romania
| | - Olga Soritau
- Department of Radiobiology and Tumor Biology, Oncology Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Cluj, Romania
| | - Andreea Campu
- Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania.
| | - Luiza Gaina
- The Research Centre on Fundamental and Applied Heterochemistry, Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania
| | - Emoke Pall
- Department of Infectious Diseases, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Cluj, Romania
| | - Bogdan Pop
- Department of Pathology, Oncology Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Cluj, Romania and Department of Pathology, University of Medicine and Pharmacy Iuliu HaŢieganu, Cluj-Napoca, Cluj, Romania
| | - Oana Baldasici
- Department of Functional Genomics, Proteomics and Experimental Pathology, Oncology Institute Prof. Dr. Ion Chiricuta, Cluj-Napoca, Cluj, Romania
| | - Claudia Gherman
- Department of Functional Genomics, Proteomics and Experimental Pathology, Oncology Institute Prof. Dr Ion Chiricuta, Cluj-Napoca, Cluj, Romania
| | - Daria Stoia
- Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania.
| | - Dana Maniu
- Biomolecular Physics Department, Faculty of Physics, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania. and Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania.
| | - Simion Astilean
- Biomolecular Physics Department, Faculty of Physics, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania. and Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Cluj, Romania.
| |
Collapse
|
34
|
Demarchi MS, Seeliger B, Lifante JC, Alesina PF, Triponez F. Fluorescence Image-Guided Surgery for Thyroid Cancer: Utility for Preventing Hypoparathyroidism. Cancers (Basel) 2021; 13:cancers13153792. [PMID: 34359693 PMCID: PMC8345196 DOI: 10.3390/cancers13153792] [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: 05/25/2021] [Revised: 06/08/2021] [Accepted: 07/23/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Hypoparathyroidism is one of the most frequent complications of thyroid surgery, especially when associated with lymph node dissection in cases of thyroid cancer. Fluorescence-guided surgery is an emerging tool that appears to help reduce the rate of this complication. The present review aims to highlight the utility of fluorescence imaging in preserving parathyroid glands during thyroid cancer surgery. Methods: We performed a systematic review of the literature according to PRISMA guidelines to identify published studies on fluorescence-guided thyroid surgery with a particular focus on thyroid cancer. Articles were selected and analyzed per indication and type of surgery, autofluorescence or exogenous dye usage, and outcomes. The Methodological Index for Non-Randomized Studies (MINORS) was used to assess the methodological quality of the included articles. Results: Twenty-five studies met the inclusion criteria, with three studies exclusively assessing patients with thyroid cancer. The remaining studies assessed mixed cohorts with thyroid cancer and other thyroid or parathyroid diseases. The majority of the papers support the potential benefit of fluorescence imaging in preserving parathyroid glands in thyroid surgery. Conclusions: Fluorescence-guided surgery is useful in the prevention of post-thyroidectomy hypoparathyroidism via enhanced early identification, visualization, and preservation of the parathyroid glands. These aspects are notably beneficial in cases of associated lymphadenectomy for thyroid cancer.
Collapse
Affiliation(s)
- Marco Stefano Demarchi
- Department of Thoracic and Endocrine Surgery and Faculty of Medicine, University Hospitals of Geneva, 4 Rue Gabrielle Perret-Gentil, 1211 Geneva, Switzerland;
- Department of Endocrine Surgery, Lyon Sud University Hospitals, 69310 Pierre Benite, France;
| | - Barbara Seeliger
- IHU—Strasbourg, Institute of Image-Guided Surgery, 67091 Strasbourg CEDEX, France;
- IRCAD, Research Institute against Digestive Cancer, 67091 Strasbourg CEDEX, France
- Department of General, Digestive, and Endocrine Surgery, Strasbourg University Hospitals, 67091 Strasbourg CEDEX, France
- Department of Surgery and Center of Minimally Invasive Surgery, Evangelische Kliniken Essen-Mitte, Academic Teaching Hospital of the University of Duisburg-Essen, 45136 Essen, Germany;
| | - Jean-Christophe Lifante
- Department of Endocrine Surgery, Lyon Sud University Hospitals, 69310 Pierre Benite, France;
- Health Services and Performance Research Lab (EA 7425 HESPER), Université Claude Bernard Lyon 1, 69622 Lyon, France
| | - Pier Francesco Alesina
- Department of Surgery and Center of Minimally Invasive Surgery, Evangelische Kliniken Essen-Mitte, Academic Teaching Hospital of the University of Duisburg-Essen, 45136 Essen, Germany;
- Department of Surgery, Gemelli Molise Hospital, Università Cattolica del Sacro Cuore, 86100 Campobasso, Italy
| | - Frédéric Triponez
- Department of Thoracic and Endocrine Surgery and Faculty of Medicine, University Hospitals of Geneva, 4 Rue Gabrielle Perret-Gentil, 1211 Geneva, Switzerland;
- Correspondence: ; Tel.: +41-(0)22-372-78-62
| |
Collapse
|
35
|
olde Heuvel J, de Wit-van der Veen BJ, Huizing DM, van der Poel HG, van Leeuwen PJ, Bhairosing PA, Stokkel MP, Slump CH. State-of-the-art Intraoperative Imaging Technologies for Prostate Margin Assessment: A Systematic Review. Eur Urol Focus 2021; 7:733-741. [DOI: 10.1016/j.euf.2020.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/06/2020] [Accepted: 02/05/2020] [Indexed: 12/29/2022]
|
36
|
Design of a Near Infrared Fluorescent Ureter Imaging Agent for Prevention of Ureter Damage during Abdominal Surgeries. Molecules 2021; 26:molecules26123739. [PMID: 34205289 PMCID: PMC8234099 DOI: 10.3390/molecules26123739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022] Open
Abstract
The inadvertent severing of a ureter during surgery occurs in as many as 4.5% of colorectal surgeries. To help prevent this issue, several near-infrared (NIR) dyes have been developed to assist surgeons with identifying ureter location. However, the majority of these dyes exhibit at least some issue that precludes their widespread usage such as high levels of uptake in other tissues, overlapping emission wavelengths with other NIR dyes used for other fluorescence-guided surgeries, and/or rapid excretion times through the ureters. To overcome these limitations, we have synthesized and characterized the spectral properties and biodistribution of a new series of PEGylated UreterGlow derivatives. The most promising dye, UreterGlow-11 was shown to almost exclusively excrete through the kidneys/ureters with detectable fluorescence observed for at least 12 h. Additionally, while the excitation wavelength is similar to that of other NIR dyes used for cancer resections, the emission is shifted by ~30 nm allowing for discrimination between the different fluorescence-guided surgery probes. In conclusion, these new UreterGlow dyes show promising optical and biodistribution characteristics and are good candidates for translation into the clinic.
Collapse
|
37
|
Alfonso-Garcia A, Bec J, Weyers B, Marsden M, Zhou X, Li C, Marcu L. Mesoscopic fluorescence lifetime imaging: Fundamental principles, clinical applications and future directions. JOURNAL OF BIOPHOTONICS 2021; 14:e202000472. [PMID: 33710785 PMCID: PMC8579869 DOI: 10.1002/jbio.202000472] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 05/16/2023]
Abstract
Fluorescence lifetime imaging (FLIm) is an optical spectroscopic imaging technique capable of real-time assessments of tissue properties in clinical settings. Label-free FLIm is sensitive to changes in tissue structure and biochemistry resulting from pathological conditions, thus providing optical contrast to identify and monitor the progression of disease. Technical and methodological advances over the last two decades have enabled the development of FLIm instrumentation for real-time, in situ, mesoscopic imaging compatible with standard clinical workflows. Herein, we review the fundamental working principles of mesoscopic FLIm, discuss the technical characteristics of current clinical FLIm instrumentation, highlight the most commonly used analytical methods to interpret fluorescence lifetime data and discuss the recent applications of FLIm in surgical oncology and cardiovascular diagnostics. Finally, we conclude with an outlook on the future directions of clinical FLIm.
Collapse
Affiliation(s)
- Alba Alfonso-Garcia
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Julien Bec
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Brent Weyers
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Mark Marsden
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Xiangnan Zhou
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Cai Li
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Laura Marcu
- Department of Biomedical Engineering, University of California, Davis, Davis, California
- Department Neurological Surgery, University of California, Davis, California
| |
Collapse
|
38
|
Inamori K, Togashi Y, Fukuoka S, Akagi K, Ogasawara K, Irie T, Motooka D, Kobayashi Y, Sugiyama D, Kojima M, Shiiya N, Nakamura S, Maruyama S, Suzuki Y, Ito M, Nishikawa H. Importance of lymph node immune responses in MSI-H/dMMR colorectal cancer. JCI Insight 2021; 6:137365. [PMID: 33755600 PMCID: PMC8262295 DOI: 10.1172/jci.insight.137365] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/18/2021] [Indexed: 01/21/2023] Open
Abstract
Patients with colorectal cancers (CRCs) generally exhibit improved survival through intensive lymph node (LN) dissection. However, recent progress in cancer immunotherapy revisits the potential importance of regional LNs, where T cells are primed to attack tumor cells. To elucidate the role of regional LN, we investigated the immunological status of nonmetastatic regional LN lymphocytes (LNLs) in comparison with those of the tumor microenvironment (tumor-infiltrating lymphocytes; TILs) using flow cytometry and next-generation sequencing. LNLs comprised an intermediate level of the effector T cell population between peripheral blood lymphocytes (PBLs) and TILs. Significant overlap of the T cell receptor (TCR) repertoire was observed in microsatellite instability–high/mismatch repair–deficient (MSI-H/dMMR) CRCs with high tumor mutation burden (TMB), although limited TCRs were shared between nonmetastatic LNs and primary tumors in microsatellite stable/MMR proficient (MSS/pMMR) CRC patients with low TMB. In line with the overlap of the TCR repertoire, an excessive LN dissection did not provide a positive impact on long-term prognosis in our MSI-H/dMMR CRC cohort (n = 130). We propose that regional LNs play an important role in antitumor immunity, particularly in MSI-H/dMMR CRCs with high TMB, requiring care to be taken regarding excessive nonmetastatic LN dissection in MSI-H/dMMR CRC patients.
Collapse
Affiliation(s)
- Koji Inamori
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan.,Department of Colorectal Surgery, National Cancer Center Hospital East (NCCHE), Chiba, Japan.,Surgery 1, Divisions of cardiovascular, Thoracic, General Endoscopic and Breast Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yosuke Togashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan
| | - Shota Fukuoka
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan
| | - Kiwamu Akagi
- Division of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center (SCC), Saitama, Japan
| | - Kouetsu Ogasawara
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Takuma Irie
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan
| | - Daisuke Motooka
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoichi Kobayashi
- Department of Immunology and.,Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | - Norihiko Shiiya
- Surgery 1, Divisions of cardiovascular, Thoracic, General Endoscopic and Breast Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shota Nakamura
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Masaaki Ito
- Department of Colorectal Surgery, National Cancer Center Hospital East (NCCHE), Chiba, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo/Chiba, Japan.,Department of Immunology and
| |
Collapse
|
39
|
Heidkamp J, Scholte M, Rosman C, Manohar S, Fütterer JJ, Rovers MM. Novel imaging techniques for intraoperative margin assessment in surgical oncology: A systematic review. Int J Cancer 2021; 149:635-645. [PMID: 33739453 PMCID: PMC8252509 DOI: 10.1002/ijc.33570] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/08/2020] [Accepted: 03/01/2021] [Indexed: 12/25/2022]
Abstract
Inadequate margins continue to occur frequently in patients who undergo surgical resection of a tumor, suggesting that current intraoperative methods are not sufficiently reliable in determining the margin status. This clinical demand has inspired the development of many novel imaging techniques that could help surgeons with intraoperative margin assessment. This systematic review provides an overview of novel imaging techniques for intraoperative margin assessment in surgical oncology, and reports on their technical properties, feasibility in clinical practice and diagnostic accuracy. PubMed, Embase, Web of Science and the Cochrane library were systematically searched (2013‐2018) for studies reporting on imaging techniques for intraoperative margin assessment. Patient and study characteristics, technical properties, feasibility characteristics and diagnostic accuracy were extracted. This systematic review identified 134 studies that investigated and developed 16 groups of techniques for intraoperative margin assessment: fluorescence, advanced microscopy, ultrasound, specimen radiography, optical coherence tomography, magnetic resonance imaging, elastic scattering spectroscopy, bio‐impedance, X‐ray computed tomography, mass spectrometry, Raman spectroscopy, nuclear medicine imaging, terahertz imaging, photoacoustic imaging, hyperspectral imaging and pH measurement. Most studies were in early developmental stages (IDEAL 1 or 2a, n = 98); high‐quality stage 2b and 3 studies were rare. None of the techniques was found to be clearly superior in demonstrating high feasibility as well as high diagnostic accuracy. In conclusion, the field of imaging techniques for intraoperative margin assessment is highly evolving. This review provides a unique overview of the opportunities and limitations of the currently available imaging techniques.
What's new?
While surgical resection is critical in the treatment of primary solid tumors, resection at tumor margins remains problematic, with inadequately resected margins facilitating tumor recurrence. In this systematic review, the authors collected information on novel imaging techniques applied to the intraoperative assessment of tumor margins across cancer types. A total of 16 groups of techniques were identified, with many in early stages of clinical application. Following comparison, no single technique was clearly superior in clinical feasibility or diagnostic accuracy. The review highlights the evolving nature of imaging techniques for intraoperative margin assessment and identifies opportunities and limitations in the field.
Collapse
Affiliation(s)
- Jan Heidkamp
- Department of Radiology and Nuclear Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mirre Scholte
- Department of Operating Rooms, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Camiel Rosman
- Department of Surgery, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Srirang Manohar
- Multi-Modality Medical Imaging group, Technical Medical Center, University of Twente, Enschede, The Netherlands
| | - Jurgen J Fütterer
- Department of Radiology and Nuclear Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maroeska M Rovers
- Department of Operating Rooms, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Health Evidence, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
40
|
Borlan R, Focsan M, Maniu D, Astilean S. Interventional NIR Fluorescence Imaging of Cancer: Review on Next Generation of Dye-Loaded Protein-Based Nanoparticles for Real-Time Feedback During Cancer Surgery. Int J Nanomedicine 2021; 16:2147-2171. [PMID: 33746512 PMCID: PMC7966856 DOI: 10.2147/ijn.s295234] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
The use of fluorescence imaging technique for visualization, resection and treatment of cancerous tissue, attained plenty of interest once the promise of whole body and deep tissue near-infrared (NIR) imaging emerged. Why is NIR so desired? Contrast agents with optical properties in the NIR spectral range offer an upgrade for the diagnosis and treatment of cancer, by dint of the deep tissue penetration of light in the NIR region of the electromagnetic spectrum, also known as the optical window in biological tissue. Thus, the development of a new generation of NIR emitting and absorbing contrast agents able to overcome the shortcomings of the basic free dye administration is absolutely essential. Several examples of nanoparticles (NPs) have been successfully implemented as carriers for NIR dye molecules to the tumour site owing to their prolonged blood circulation time and enhanced accumulation within the tumour, as well as their increased fluorescence signal relative to free fluorophore emission and active targeting of cancerous cells. Due to their versatile structure, good biocompatibility and capability to efficiently load dyes and bioconjugate with diverse cancer-targeting ligands, the research area of developing protein-based NPs encapsulated or conjugated with NIR dyes is highly promising but still in its infancy. The current review aims to provide an up-to-date overview on the biocompatibility, specific targeting and versatility offered by protein-based NPs loaded with different classes of NIR dyes as next-generation fluorescent agents. Moreover, this study brings to light the newest and most relevant advances involving the state-of-the-art NIR fluorescent agents for the real-time interventional NIR fluorescence imaging of cancer in clinical trials.
Collapse
Affiliation(s)
- Raluca Borlan
- Biomolecular Physics Department, Faculty of Physics, Babeș-Bolyai University, Cluj-Napoca, Cluj, Romania.,Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babeș-Bolyai University, Cluj-Napoca, Cluj, Romania
| | - Monica Focsan
- Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babeș-Bolyai University, Cluj-Napoca, Cluj, Romania
| | - Dana Maniu
- Biomolecular Physics Department, Faculty of Physics, Babeș-Bolyai University, Cluj-Napoca, Cluj, Romania
| | - Simion Astilean
- Biomolecular Physics Department, Faculty of Physics, Babeș-Bolyai University, Cluj-Napoca, Cluj, Romania.,Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute in Bio-Nano-Sciences, Babeș-Bolyai University, Cluj-Napoca, Cluj, Romania
| |
Collapse
|
41
|
de Boer NL, Brandt-Kerkhof ARM, Madsen EVE, Doukas M, Verhoef C, Burger JWA. The Accuracy of the Surgical Peritoneal Cancer Index in Patients with Peritoneal Metastases of Colorectal Cancer. Dig Surg 2021; 38:205-211. [PMID: 33657551 DOI: 10.1159/000513353] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 11/15/2020] [Indexed: 12/10/2022]
Abstract
INTRODUCTION The peritoneal cancer index (PCI) is one of the most important prognostic factors in patients with peritoneal metastases from colorectal cancer undergoing cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS-HIPEC). The PCI is determined during laparotomy by 2 experienced surgeons and plays a major role in the decision to proceed with CRS-HIPEC. The primary objective of this study was to determine the accuracy of the surgical PCI (sPCI) by comparing it with the PCI confirmed by the pathologist (pPCI). METHODS All consecutive patients who underwent CRS-HIPEC for colorectal peritoneal metastases between February 2015 and June 2018 were identified. Relevant patient- and tumor-related characteristics were collected. RESULTS In total, 119 patients were included, 60 males (50.4%). The median age was 64 (IQR 55-71). The median sPCI (sPCI = 11, IQR 6-16) was significantly higher than the median pPCI (pPCI = 8, IQR 3-13, p < 0.001). The total pPCI was lower than the total sPCI in 80 patients (67.2%). In 21 patients (17.6%), the sPCI was overestimated with ≥5 points. Small lesions are more likely to be negative. In patients that underwent resection of their primary tumor prior to CRS-HIPEC, the difference between the sPCI and pPCI was significantly larger (p < 0.05). CONCLUSIONS Surgical calculation of the PCI often results in overestimation. Far-reaching consequences are tied to the macroscopic evaluation of the sPCI, but this evaluation seems not very reliable.
Collapse
Affiliation(s)
- Nadine L de Boer
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands,
| | | | - Eva V E Madsen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Michael Doukas
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jacobus W A Burger
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.,Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| |
Collapse
|
42
|
Lee YJ, Krishnan G, Nishio N, van den Berg NS, Lu G, Martin BA, van Keulen S, Colevas AD, Kapoor S, Liu JTC, Rosenthal EL. Intraoperative Fluorescence-Guided Surgery in Head and Neck Squamous Cell Carcinoma. Laryngoscope 2021; 131:529-534. [PMID: 33593036 DOI: 10.1002/lary.28822] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023]
Abstract
The rate of positive margins in head and neck cancers has remained stagnant over the past three decades and is consistently associated with poor overall survival. This suggests that significant improvements must be made intraoperatively to ensure negative margins. We discuss the important role of fluorescence imaging to guide surgical oncology in head and neck cancer. This review includes a general overview of the principles of fluorescence, available fluorophores used for fluorescence imaging, and specific clinical applications of fluorescence-guided surgery, as well as challenges and future directions in head and neck surgical oncology. Laryngoscope, 131:529-534, 2021.
Collapse
Affiliation(s)
- Yu-Jin Lee
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Giri Krishnan
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A.,Department of Otolaryngology, Head and Neck Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Naoki Nishio
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Nynke S van den Berg
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Guolan Lu
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Brock A Martin
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Stan van Keulen
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Alexander D Colevas
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Shrey Kapoor
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Jonathan T C Liu
- Department of Mechanical Engineering, University of Washington, Seattle, WA, U.S.A.,Department of Bioengineering, University of Washington, Seattle, WA, U.S.A.,Department of Pathology, University of Washington, Seattle, WA, U.S.A
| | - Eben L Rosenthal
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A.,Department of Radiology, Stanford University School of Medicine, Stanford, CA, U.S.A
| |
Collapse
|
43
|
Functional Imaging Using Bioluminescent Reporter Genes in Living Subjects. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
44
|
Xing J, Gong Q, Akakuru OU, Liu C, Zou R, Wu A. Research advances in integrated theranostic probes for tumor fluorescence visualization and treatment. NANOSCALE 2020; 12:24311-24330. [PMID: 33300527 DOI: 10.1039/d0nr06867e] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
At present, cancer is obviously a major threat to human health worldwide. Accurate diagnosis and treatment are in great demand and have become an effective method to alleviate the development of cancer and improve the survival rate of patients. A large number of theranostic probes that combine diagnosis and treatment methods have been developed as promising tools for tumor precision medicine. Among them, fluorescent theranostic probes have developed rapidly in the frontier research field of precision medicine with their real time, low toxicity, and high-resolution merit. Therefore, this review focuses on recent advances in the development of fluorescent theranostic probes, as well as their applications for cancer diagnosis and treatment. Initially, small-molecule fluorescent theranostic probes mainly including tumor microenvironment-responsive fluorescent prodrugs and phototherapeutic probes were introduced. Subsequently, nanocomposite probes are expounded based on four types of nano-fluorescent particles combining different therapies (chemotherapy, photothermal therapy, photodynamic therapy, gene therapy, etc.). Then, the capsule-type "all in one" probes, which occupy an important position in theranostic probes, are summarized according to the surface carrier type. This review aims to present a comprehensive guide for researchers in the field of tumor-related theranostic probe design and development.
Collapse
Affiliation(s)
- Jie Xing
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China. and University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Qiuyu Gong
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China.
| | - Ozioma Udochukwu Akakuru
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China. and University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Chuang Liu
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China. and University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Ruifen Zou
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China.
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, PR China.
| |
Collapse
|
45
|
Ji Y, Jones C, Baek Y, Park GK, Kashiwagi S, Choi HS. Near-infrared fluorescence imaging in immunotherapy. Adv Drug Deliv Rev 2020; 167:121-134. [PMID: 32579891 DOI: 10.1016/j.addr.2020.06.012] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
Near-infrared (NIR) light possesses many suitable optophysical properties for medical imaging including low autofluorescence, deep tissue penetration, and minimal light scattering, which together allow for high-resolution imaging of biological tissue. NIR imaging has proven to be a noninvasive and effective real-time imaging methodology that provides a high signal-to-background ratio compared to other potential optical imaging modalities. In response to this, the use of NIR imaging has been extensively explored in the field of immunotherapy. To date, NIR fluorescence imaging has successfully offered reliable monitoring of the localization, dynamics, and function of immune responses, which are vital in assessing not only the efficacy but also the safety of treatments to design immunotherapies optimally. This review aims to provide an overview of the current research on NIR imaging of the immune response. We expect that the use of NIR imaging will expand further in response to the recent success in cancer immunotherapy. We will also offer our insights on how this technology will meet rapidly growing expectations in the future.
Collapse
Affiliation(s)
- Yuanyuan Ji
- Scientific Research Centre, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China; Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Catherine Jones
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yoonji Baek
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - G Kate Park
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
46
|
Turan MI, Celik M, Ertürk MS. Indocyanine green fluorescence angiography-guided transoral endoscopic thyroidectomy and parathyroidectomy: First clinical report. Photodiagnosis Photodyn Ther 2020; 32:102028. [PMID: 32979545 DOI: 10.1016/j.pdpdt.2020.102028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/05/2020] [Accepted: 09/14/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Indocyanine green fluorescence (ICG) angiography has been used for many purposes including as part of a focused parathyroidectomy technique. Concomitant fluorescence of thyroid tissue may cause challenges defining parathyroid tissue during surgery, since ICG is not a selective fluorescent agent. On the other hand, cosmesis is still a big problem for patients due to the visible neck scars produced by the standard surgical technique. In this study, we described a novel technique to solve both these handicaps. MATERIALS AND METHODS Seven patients who underwent ICG fluorescence angiography-guided transoral endoscopic thyroidectomy and parathyroidectomy vestibular approach between February 2018 and July 2019 were included. Serum parathyroid hormone (PTH) levels were measured intraoperatively and on postoperative day 1. Fluorescent images were confirmed with intraoperative quick-PTH levels. RESULTS All operations were done successfully without conversion to open surgery. Intense and isolated parathyroid fluorescent images were achieved in all operations. All patients had a 50 % decrease between the baseline and final quick-PTH levels and the final quick-PTH levels were in the normal range in all. One of 7 patients had epistaxis due to nasotracheal intubation. One of 7 patients had seroma on post-operative day 5. None of patients had mental nerve injury, permanent hypocalcemia and temporary or permanent recurrent laryngeal nerve injury. CONCLUSION ICG-guided transoral endoscopic thyroid and parathyroid surgery can be used in select patients to increase operative success in focused parathyroidectomy with excellent cosmetic outcome also noted.
Collapse
Affiliation(s)
| | - Mehmet Celik
- Trakya University Medical Faculty, Department of Endocrinology and Metabolism, Edirne, Turkey
| | - Mehmet Sercan Ertürk
- University of Health Sciences Antalya Training and Research Hospital, Department of Endocrinology and Metabolism, Antalya, Turkey
| |
Collapse
|
47
|
Borlan R, Tatar AS, Soritau O, Maniu D, Marc G, Florea A, Focsan M, Astilean S. Design of fluorophore-loaded human serum albumin nanoparticles for specific targeting of NIH:OVCAR3 ovarian cancer cells. NANOTECHNOLOGY 2020; 31:315102. [PMID: 32315999 DOI: 10.1088/1361-6528/ab8b90] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nowadays, extensive research is being carried out to find innovative solutions for the development of stable, reproductible, and highly efficient fluorescent contrast agents with the ability of targeting specific cells, which can be further implemented for fluorescent-guided surgery in a real clinical setting. The present study is focused on the development of fluorescent dye-loaded protein nanoparticles (NPs) to overcome the drawbacks of the standard administration of free organic fluorophores, such as cytotoxicity, aqueousinstability, and rapid photo-degradation. Precisely, human serum albumin (HSA) NPs loaded with two different FDA approved dyes, namely indocyanine green (ICG) and fluorescein isothiocyanate (FITC), with a fluorescence response in the near-infrared and visible spectral domains, respectively, have been successfully designed. Even though the diameter of fluorescent HSA NPs is around 30 nm as proven by dynamic light scattering and transmission electron microscopy investigations, they present good loading efficiencies of almost 50% for ICG, and over 30% for FITC and a high particle yield of over 75%. Molecular docking simulations of ICG and FITC within the structure of HSA confirmed that the dyes were loaded inside the NPs, and docked in Site I (subdomain IIA) of the HSA molecule. After the confirmation of their high fluorescence photostability, the NPs were covalently conjugated with folic acid (HSA-FA NPs) in order to bind specifically to the folate receptor alpha (FRα) protein overexpressed on NIH:OVCAR3 ovarian cancer cells. Finally, fluorescence microscopy imaging investigations validate the improved internalization of folate targeted HSA&FITC NPs compared to cells treated with untargeted ones. Furthermore, TEM examinations of the distribution of HSA NPs into the NIH:OVCAR3 cells revealed anincreased number of NP-containing vesicles for the cells treated with HSA-FA NPs, compared to the cells exposed to untargeted HAS NPs, upholding the enhanced cellular uptake through FRα-mediated potocytosis.
Collapse
Affiliation(s)
- Raluca Borlan
- Biomolecular Physics Department, Faculty of Physics, Babeş-Bolyai University, 1 Kogălniceanu Street, Cluj-Napoca 400084, Romania. Nanobiophotonics and Laser Microspectroscopy Centre, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babeş-Bolyai University, 42 Treboniu Laurean Street, Cluj-Napoca 400271, Romania
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Baiocchi GL, Gheza F, Molfino S, Arru L, Vaira M, Giacopuzzi S. Indocyanine green fluorescence-guided intraoperative detection of peritoneal carcinomatosis: systematic review. BMC Surg 2020; 20:158. [PMID: 32680492 PMCID: PMC7367360 DOI: 10.1186/s12893-020-00821-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/08/2020] [Indexed: 02/08/2023] Open
Abstract
Background To review the available clinical data about the value of Indocyanine Green (ICG) fluorescence imaging for intraoperative detection of peritoneal carcinomatosis. Methods We conducted a systematic review, according to the PRISMA guidelines, for clinical series investigating the possible role of ICG fluorescence imaging in detecting peritoneal carcinomatosis during surgical treatment of abdominal malignancies. With the aim to analyze actual application in the daily clinical practice, papers including trials with fluorophores other than ICG, in vitro and animals series were excluded. Data on patients and cancer features, timing, dose and modality of ICG administration, sensitivity, specificity and accuracy of fluorescence diagnosis of peritoneal nodules were extracted and analyzed. Results Out of 192 screened papers, we finally retrieved 7 series reporting ICG-guided detection of peritoneal carcinomatosis. Two papers reported the same cases, thus only 6 series were analyzed, for a total of 71 patients and 353 peritoneal nodules. The investigated tumors were colorectal carcinomas in 28 cases, hepatocellular carcinoma in 16 cases, ovarian cancer in 26 cases and endometrial cancer in 1 case. In all but 4 cases, the clinical setting was an elective intervention in patients known as having peritoneal carcinomatosis. No series reported a laparoscopic procedure. Technical data of ICG management were consistent across the studies. Overall, 353 lesions were harvested and singularly evaluated. Sensitivity varied from 72.4 to 100%, specificity from 54.2 to 100%. Two series reported that planned intervention changed in 25 and 29% of patients, respectively. Conclusion Indocyanine Green based fluorescence of peritoneal carcinomatosis is a promising intraoperative tool for detection and characterization of peritoneal nodules in patients with colorectal, hepatocellular, ovarian carcinomas. Further prospective studies are needed to fix its actual diagnostic value on these and other abdominal malignancies with frequent spread to peritoneum.
Collapse
Affiliation(s)
- Gian Luca Baiocchi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy. .,Oncoteam Peritoneal Surface Malignancies, SICO (Italian Society of Surgical Oncology), Brescia, Italy.
| | - Federico Gheza
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Sarah Molfino
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Luca Arru
- Centre Hospitalier de Luxembourg, Service de Chirurgie Generale, Luxemborg City, Luxembourg
| | - Marco Vaira
- Oncoteam Peritoneal Surface Malignancies, SICO (Italian Society of Surgical Oncology), Brescia, Italy.,Surgical Oncology Unit, Candiolo Cancer Institute, FPO - IRCCS, Str. Prov.le 142, km. 3,95, 10060, Candiolo, TO, Italy
| | - Simone Giacopuzzi
- Department of Surgery, General and Upper G.I. Surgery Division, University of Verona, Verona, Italy
| |
Collapse
|
49
|
Zheng S, Zhang Y, Chen S, Zhang Z, Chen F, Zhang Z, Hu Z, Tian J, Wang L. A preliminary study of dual-band confocal laser endomicroscopy combined with image mosaic in the diagnosis of liver cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102250. [PMID: 32619706 DOI: 10.1016/j.nano.2020.102250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/20/2022]
Abstract
Accurate identification of tumor tissues and their margins are still challenging for conventional clinical imaging methods during liver cancer surgery. In this study, dual-band confocal laser endomicroscopy (CLE) combined with image mosaic was used to guide liver cancer surgery. In the experiments with mice bearing orthotropic liver tumor, CLE can accurately detect the tumors and identify their margins with two excitation wavelengths of 488 nm and 660 nm by clinically available dyes fluorescein sodium (FS) or indocyanine green (ICG). The mosaic CLE images enlarged the imaging field and detected the liver tumor margins more accurately. Normal liver tissues fluorescence intensity of CLE images was significantly higher than that of tumor tissues in the same tumor-bearing mice (P < 0.0001). Overall, dual-band CLE imaging demonstrates to be a promising method to identify liver tumor tissues and margins, which has the prospect of clinical application and helps to achieve intraoperative radical resection.
Collapse
Affiliation(s)
- Sheng Zheng
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ying Zhang
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China; Department of Gastroenterology, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Shujie Chen
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China; Department of Gastroenterology, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Zeyu Zhang
- 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; University of Chinese Academy of Sciences, Beijing, China
| | - Fei Chen
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Zizhen Zhang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, 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; University of Chinese Academy of Sciences, Beijing, China.
| | - Liangjing Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
50
|
Newton A, Predina J, Mison M, Runge J, Bradley C, Stefanovski D, Singhal S, Holt D. Intraoperative near-infrared imaging can identify canine mammary tumors, a spontaneously occurring, large animal model of human breast cancer. PLoS One 2020; 15:e0234791. [PMID: 32555698 PMCID: PMC7299356 DOI: 10.1371/journal.pone.0234791] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/02/2020] [Indexed: 01/05/2023] Open
Abstract
Introduction Current methods of intraoperative margin assessment in breast conserving surgery are impractical, unreliable, or time consuming. We hypothesized that intraoperative near-infrared (NIR) imaging with an FDA-approved NIR optical contrast agent could identify canine mammary tumors, a spontaneous large animal model of human breast cancer, during surgery. Methods Dogs with mammary tumors underwent a standard of care lumpectomy or mastectomy with wide surgical margins 20 hours after indocyanine green administration (3 mg/kg IV). During surgery, NIR imaging was performed on tumors and wound margins in situ and tumors and lymph nodes ex vivo. Following resection, the wound bed was examined for residual fluorescence. Fluorescence intensity was determined by signal-to-background ratio (SBR). All tumors, areas of residual fluorescence, and lymph nodes underwent histopathologic analysis. Results There were 41 mammary tumors in 16 female dogs. Twenty tumors were malignant and 21 were benign. Twenty-eight tumors were fluorescent (mean SBR 1.5±0.2). Sensitivity of fluorescence for all malignant tumors was 80% (16/20) and 93.3% (14/15) for malignant tumors > 2 cm. Specificity for malignancy was low (< 2cm = 55%; > 2cm = 30%). Tumors > 2 cm were more likely to be fluorescent (OR 6.05, 95% CI 1.50–24.44, P = 0.011) but not more likely to be malignant (OR 3.09, 95% CI 0.86–11.14, P = 0.085) than tumors ≤ 2 cm. Four out of seven inguinal lymph nodes excised in the mastectomy specimen fluoresced. All four drained malignant tumors; however only 2/4 contained metastatic disease. Conclusion Systemic ICG accumulates reliably in malignant canine mammary tumors > 2 cm. Although no tumor margins fluoresced, a wider margin of normal tissue is removed in canine mastectomy, making direct comparisons with breast conserving surgery difficult. Targeted NIR imaging agents are likely required to improve detection of smaller tumors and improve the specificity of NIR imaging for residual disease and metastatic lymph node detection.
Collapse
Affiliation(s)
- Andrew Newton
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jarrod Predina
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michael Mison
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jeffrey Runge
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Charles Bradley
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Darko Stefanovski
- Department of Clinical Studies New Bolton Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - David Holt
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|