51
|
Lwin TM, Hernot S, Hollandsworth H, Amirfakhri S, Filemoni F, Debie P, Hoffman RM, Bouvet M. Tumor-specific near-infrared nanobody probe rapidly labels tumors in an orthotopic mouse model of pancreatic cancer. Surgery 2020; 168:85-91. [PMID: 32370916 DOI: 10.1016/j.surg.2020.02.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/07/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Nanobodies, derived from camelid antibodies made of only heavy chains, are the smallest, biologic, antigen-binding fragments (~15kDa) with faster pharmacokinetics and better tumor penetration efficiency than standard antibodies. The present study evaluates the efficacy of a fluorescent, anti-carcinoembryonic antigen (CEA) nanobody for rapid tumor labeling in an orthotopic mouse model of pancreatic cancer. METHODS Anti-CEA or control nanobodies were conjugated with the near-infrared fluorophore IRDye 800CW. Fragments of BxPC-3 (high-CEA expressing) or MiaPACA-2 (low-CEA expressing) human pancreatic cancer cell lines were orthotopically implanted into the pancreatic tail of nude mice. After tumors reached 7 to 10 mm in size, 2 nmol anti-CEA or control nanobody-IRDye800CW were injected intravenously. Mice were imaged at various time points hours post-injection. RESULTS Anti-CEA nanobodies clearly labeled BxPC3 orthotopic pancreatic tumors 3 hours after injection. The signal was present as early as 15 minutes after injection and was robust at 1 to 3 hours after injection with a tumor-to-background ratio of 2.66. In contrast, there was very low accumulation in the low CEA-expressing, MiaPACA2 pancreatic orthotopic tumors. The fluorophore-conjugated nanobody was specific for CEA-expressing tumors, while the control nanobody did not show any tumor-specific signal. Both nanobodies had strong kidney uptake as expected for small-molecule probes. The fluorescence signal was detectable using 2 clinical, Food and Drug Administration-approved, 800 nm imaging devices as well as small animal imaging systems. CONCLUSION This anti-CEA, nanobody-based, fluorescent probe labeled pancreatic orthotopic tumors within 15 minutes of intravenous injection. Fluorescent anti-CEA nanobodies have labeling kinetics that approach the speed of nonspecific dyes such as indocyanine green but with the specificity of antibodies. The use of fluorescently-labeled, intact antibodies leads to a labeling delay of 48 to 96 hours between probe administration and the necessarily delayed time of operation, which can be avoided with nanobodies. The kinetics of a nanobody-based probe makes it a practical agent for same-day, patient administration and fluorescence-guided surgery.
Collapse
Affiliation(s)
- Thinzar M Lwin
- Department of Surgery, University of California San Diego, CA
| | - Sophie Hernot
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hannah Hollandsworth
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA
| | - Filemoni Filemoni
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA
| | - Pieterjan Debie
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA; AntiCancer, Inc, San Diego, CA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA.
| |
Collapse
|
52
|
Zheng Y, Hou G, Zhang G, Lan T, Yuan J, Zhang L, Yan F, Wang F, Meng P, Dun X, Li X, Chen G, Zhu Z, Wei D, He W, Yuan J. The near-infrared fluorescent dye IR-780 was coupled with cabazitaxel for castration-resistant prostate cancer imaging and therapy. Invest New Drugs 2020; 38:1641-1652. [DOI: 10.1007/s10637-020-00934-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023]
|
53
|
Favril S, Abma E, Stock E, Devriendt N, Van Goethem B, Blasi F, Brioschi C, Polis I, De Cock H, Miragoli L, Oliva P, Valbusa G, Vanderperren K, de Rooster H. Fluorescence-guided surgery using indocyanine green in dogs with superficial solid tumours. Vet Rec 2020; 187:273. [PMID: 32345608 DOI: 10.1136/vr.105554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 04/01/2020] [Accepted: 04/05/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Near-infrared fluorescence (NIRF) imaging is a relatively novel technique that can aid surgeons during intraoperative tumour identification. METHODS Nine canine oncology patients (five mammary gland tumours, three mast cell tumours and one melanoma) received intravenous indocyanine green (ICG). After 24 hours, tumours were resected and fluorescence intensities of tumours and surroundings were evaluated. Additional wound bed tissue was resected if residual fluorescence was present after tumour resection. Ex vivo, fluorescence-guided dissection was performed to separate tumour from surrounding tissue. RESULTS Intraoperative NIRF-guided tumour delineation was feasible in four out of nine dogs. Wound bed imaging after tumour removal identified nine additional fluorescent lesions, of which four contained tumour tissue. One of these four true positive in vivo lesions was missed by standard-of-care inspection. Ex vivo fluorescence-guided tumour dissection showed a sensitivity of 72 per cent and a specificity of 80 per cent in discriminating between tumour and surrounding tissue. CONCLUSION The value of ICG for intraoperative tumour delineation seems more limited than originally thought. Although NIRF imaging using ICG did identify remaining tumour tissue in the wound bed, a high false positive rate was also observed.
Collapse
Affiliation(s)
- Sophie Favril
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium .,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| | - Eline Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| | - Emmelie Stock
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Nausikaa Devriendt
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bart Van Goethem
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | - Ingeborgh Polis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | - Paolo Oliva
- Bracco Imaging SpA, Colleretto Giacosa, Italy
| | | | - Katrien Vanderperren
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hilde de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| |
Collapse
|
54
|
Demarchi MS, Karenovics W, Bédat B, Triponez F. Intraoperative Autofluorescence and Indocyanine Green Angiography for the Detection and Preservation of Parathyroid Glands. J Clin Med 2020; 9:jcm9030830. [PMID: 32197500 PMCID: PMC7141218 DOI: 10.3390/jcm9030830] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/01/2020] [Accepted: 03/12/2020] [Indexed: 11/16/2022] Open
Abstract
Fluorescence imaging is a well-known method for both the in vivo and in vitro identification of specific cells or tissues. This imaging tool is gaining importance in the intraoperative detection and preservation of parathyroid glands during endocrine surgery owing to the intrinsic properties of parathyroid tissue. The aim of this paper is to provide an overview of the basics of the technology, its history, and the recent surgical intraoperative applications of near-infrared imaging methods. Moreover, a literature review of the utilization of fluorescence devices in thyroid surgery suggests that the use of near-infrared imaging seems to be beneficial in reducing postoperative hypoparathyroidism, which is one of the most frequent complications of thyroid surgery.
Collapse
|
55
|
Vonk J, de Wit JG, Voskuil FJ, Witjes MJH. Improving oral cavity cancer diagnosis and treatment with fluorescence molecular imaging. Oral Dis 2020; 27:21-26. [PMID: 32072691 PMCID: PMC7818506 DOI: 10.1111/odi.13308] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/22/2022]
Abstract
Early diagnosis and radical surgical excision of oral squamous cell carcinomas are essential for achieving optimal treatment outcomes. To date, diagnostic tools that rely on anatomical anomalies provide limited information and resolution in clinical practice. As a result, oral cancer is often detected in an advanced stage. Also, no reliable real-time intraoperative tools are readily available for the evaluation of surgical resection margins. Fluorescence imaging visualises biological processes that occur in early carcinogenesis and could, therefore, enable detection of small tumours in early stages. Furthermore, due to the high sensitivity and spatial resolution, fluorescence imaging could assist in resection margin assessment during surgery. In this review, we discuss several techniques that employ fluorescence for early diagnosis and surgical guidance in oral squamous cell carcinoma and present future perspectives on the potential of fluorescence imaging in oral cancer in the near future.
Collapse
Affiliation(s)
- Jasper Vonk
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jaron Gérard de Wit
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Floris Jan Voskuil
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Max Johannes Hendrikus Witjes
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
56
|
Qu Z, Shen J, Li Q, Xu F, Wang F, Zhang X, Fan C. Near-IR emissive rare-earth nanoparticles for guided surgery. Theranostics 2020; 10:2631-2644. [PMID: 32194825 PMCID: PMC7052904 DOI: 10.7150/thno.40808] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022] Open
Abstract
Intraoperative image-guided surgery (IGS) has attracted extensive research interests in determination of tumor margins from surrounding normal tissues. Introduction of near infrared (NIR) fluorophores into IGS could significantly improve the in vivo imaging quality thus benefit IGS. Among the reported NIR fluorophores, rare-earth nanoparticles exhibit unparalleled advantages in disease theranostics by taking advantages such as large Stokes shift, sharp emission spectra, and high chemical/photochemical stability. The recent advances in elements doping and morphologies controlling endow the rare-earth nanoparticles with intriguing optical properties, including emission span to NIR-II region and long life-time photoluminescence. Particularly, NIR emissive rare earth nanoparticles hold advantages in reduction of light scattering, photon absorption and autofluorescence, largely improve the performance of nanoparticles in biological and pre-clinical applications. In this review, we systematically compared the benefits of RE nanoparticles with other NIR probes, and summarized the recent advances of NIR emissive RE nanoparticles in bioimaging, photodynamic therapy, drug delivery and NIR fluorescent IGS. The future challenges and promises of NIR emissive RE nanoparticles for IGS were also discussed.
Collapse
Affiliation(s)
- Zhibei Qu
- Joint Research Center for Precision Medicine, Shanghai Jiao Tong University & Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianlei Shen
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Feng Xu
- Joint Research Center for Precision Medicine, Shanghai Jiao Tong University & Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Fei Wang
- Joint Research Center for Precision Medicine, Shanghai Jiao Tong University & Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xueli Zhang
- Joint Research Center for Precision Medicine, Shanghai Jiao Tong University & Affiliated Sixth People's Hospital South Campus, Southern Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
57
|
Oberheim Bush NA, Hervey-Jumper SL, Berger MS. Management of Glioblastoma, Present and Future. World Neurosurg 2020; 131:328-338. [PMID: 31658576 DOI: 10.1016/j.wneu.2019.07.044] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 01/22/2023]
Abstract
Glioblastomas are the most common malignant brain tumor and despite extensive research have a dismal prognosis. This review focuses on the current treatment paradigms of glioblastoma and highlights current advances in surgical approaches, imaging techniques, molecular diagnostics, and translational efforts. Several promising clinical trials in immunotherapy and personalized medicine are discussed and the importance of quality of life in the patients and their caregivers both during active treatment and survivorship is also commented on.
Collapse
Affiliation(s)
- Nancy Ann Oberheim Bush
- Division of Neuro-Oncology, Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Shawn L Hervey-Jumper
- Division of Neuro-Oncology, Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California, USA.
| |
Collapse
|
58
|
Boss M, Bos D, Frielink C, Sandker G, Ekim S, Marciniak C, Pattou F, van Dam G, van Lith S, Brom M, Gotthardt M, Buitinga M. Targeted Optical Imaging of the Glucagonlike Peptide 1 Receptor Using Exendin-4-IRDye 800CW. J Nucl Med 2020; 61:1066-1071. [PMID: 31924726 PMCID: PMC7383075 DOI: 10.2967/jnumed.119.234542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/13/2019] [Indexed: 01/10/2023] Open
Abstract
The treatment of choice for insulinomas and focal lesions in congenital hyperinsulinism (CHI) is surgery. However, intraoperative detection can be challenging. This challenge could be overcome with intraoperative fluorescence imaging, which provides real-time lesion detection with a high spatial resolution. Here, a novel method for targeted near-infrared (NIR) fluorescence imaging of glucagonlike peptide 1 receptor (GLP-1R)–positive lesions, using the GLP-1 agonist exendin-4 labeled with IRDye 800CW, was examined in vitro and in vivo. Methods: A competitive binding assay was performed using Chinese hamster lung (CHL) cells transfected with GLP-1R. Tracer biodistribution was determined in BALB/c nude mice bearing subcutaneous CHL-GLP-1R xenografts. In vivo NIR fluorescence imaging of CHL-GLP-1R xenografts was performed. Localization of the tracer in the pancreatic islets of BALB/c nude mice was examined using fluorescence microscopy. Laparoscopic imaging was performed to detect the fluorescent signal of the tracer in the pancreas of mini pigs. Results: Exendin-4-IRDye 800CW binds GLP-1R with a half-maximal inhibitory concentration of 3.96 nM. The tracer accumulates in CHL-GLP-1R xenografts. Subcutaneous CHL-GLP-1R xenografts were visualized using in vivo NIR fluorescence imaging. The tracer accumulates specifically in the pancreatic islets of mice, and a clear fluorescent signal was detected in the pancreas of mini pigs. Conclusion: These data provide the first in vivo evidence of the feasibility of targeted fluorescence imaging of GLP-1R–positive lesions. Intraoperative lesion delineation using exendin-4-IRDye 800CW could benefit open as well as laparoscopic surgical procedures for removal of insulinomas and focal lesions in CHI.
Collapse
Affiliation(s)
- Marti Boss
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Desiree Bos
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cathelijne Frielink
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerwin Sandker
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Selen Ekim
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Camille Marciniak
- Department of General and Endocrine Surgery, University Hospital 2 Lille, Lille, France
| | - Francois Pattou
- Department of General and Endocrine Surgery, University Hospital 2 Lille, Lille, France
| | - Go van Dam
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands; and
| | - Sanne van Lith
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maarten Brom
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mijke Buitinga
- Department of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
59
|
Rondon A, Degoul F. Antibody Pretargeting Based on Bioorthogonal Click Chemistry for Cancer Imaging and Targeted Radionuclide Therapy. Bioconjug Chem 2020; 31:159-173. [PMID: 31855602 DOI: 10.1021/acs.bioconjchem.9b00761] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bioorthogonal click chemistry-employing antibody-conjugated trans-cyclooctenes (TCO) and tetrazine (Tz)-based radioligands able to covalently bind in vivo-appeared recently as a potential alternative to circumvent the hematotoxicity induced by radioimmunotherapy of solid tumors. This Review focuses on the recent advances concerning TCO/Tz pretargeting in both cancer imaging and targeted-radionuclide therapy for prospective clinical transfer. We exhaustively identified 25 PubMed publications reporting preclinical imaging and 5 therapy studies with full mAbs as targeting vectors, since its first application in 2010. The fast, safe, modulable, and specific TCO/Tz pretargeting showed high potential as a theranostic tool to get more personalized and precise cancer care. The recent optimizations reported here highlighted a possible first clinical evaluation of IEDDA pretargeting in the coming years.
Collapse
Affiliation(s)
- Aurélie Rondon
- Université Clermont Auvergne , Imagerie Moléculaire et Stratégies Théranostiques , BP 184, F-63005 Clermont-Ferrand , France.,Inserm, U 1240 , F-63000 Clermont-Ferrand , France.,Centre Jean Perrin , F-63011 Clermont-Ferrand , France
| | - Françoise Degoul
- Université Clermont Auvergne , Imagerie Moléculaire et Stratégies Théranostiques , BP 184, F-63005 Clermont-Ferrand , France.,Inserm, U 1240 , F-63000 Clermont-Ferrand , France.,Centre Jean Perrin , F-63011 Clermont-Ferrand , France
| |
Collapse
|
60
|
Najafiaghdam H, Papageorgiou E, Torquato NA, Tian B, Cohen BE, Anwar M. A 25 micron-thin microscope for imaging upconverting nanoparticles with NIR-I and NIR-II illumination. Theranostics 2019; 9:8239-8252. [PMID: 31754393 PMCID: PMC6857055 DOI: 10.7150/thno.37672] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
Rationale: Intraoperative visualization in small surgical cavities and hard-to-access areas are essential requirements for modern, minimally invasive surgeries and demand significant miniaturization. However, current optical imagers require multiple hard-to-miniaturize components including lenses, filters and optical fibers. These components restrict both the form-factor and maneuverability of these imagers, and imagers largely remain stand-alone devices with centimeter-scale dimensions. Methods: We have engineered INSITE (Immunotargeted Nanoparticle Single-Chip Imaging Technology), which integrates the unique optical properties of lanthanide-based alloyed upconverting nanoparticles (aUCNPs) with the time-resolved imaging of a 25-micron thin CMOS-based (complementary metal oxide semiconductor) imager. We have synthesized core/shell aUCNPs of different compositions and imaged their visible emission with INSITE under either NIR-I and NIR-II photoexcitation. We characterized aUCNP imaging with INSITE across both varying aUCNP composition and 980 nm and 1550 nm excitation wavelengths. To demonstrate clinical experimental validity, we also conducted an intratumoral injection into LNCaP prostate tumors in a male nude mouse that was subsequently excised and imaged with INSITE. Results: Under the low illumination fluences compatible with live animal imaging, we measure aUCNP radiative lifetimes of 600 μs - 1.3 ms, which provides strong signal for time-resolved INSITE imaging. Core/shell NaEr0.6Yb0.4F4 aUCNPs show the highest INSITE signal when illuminated at either 980 nm or 1550 nm, with signal from NIR-I excitation about an order of magnitude brighter than from NIR-II excitation. The 55 μm spatial resolution achievable with this approach is demonstrated through imaging of aUCNPs in PDMS (polydimethylsiloxane) micro-wells, showing resolution of micrometer-scale targets with single-pixel precision. INSITE imaging of intratumoral NaEr0.8Yb0.2F4 aUCNPs shows a signal-to-background ratio of 9, limited only by photodiode dark current and electronic noise. Conclusion: This work demonstrates INSITE imaging of aUCNPs in tumors, achieving an imaging platform that is thinned to just a 25 μm-thin, planar form-factor, with both NIR-I and NIR-II excitation. Based on a highly paralleled array structure INSITE is scalable, enabling direct coupling with a wide array of surgical and robotic tools for seamless integration with tissue actuation, resection or ablation.
Collapse
Affiliation(s)
- Hossein Najafiaghdam
- Department of Electrical Engineering and Computer Sciences, University of California Berkeley, Berkeley CA
| | - Efthymios Papageorgiou
- Department of Electrical Engineering and Computer Sciences, University of California Berkeley, Berkeley CA
| | - Nicole A Torquato
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Bining Tian
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Bruce E Cohen
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Mekhail Anwar
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA
| |
Collapse
|
61
|
Zheng S, Zhang Z, Qu Y, Zhang X, Guo H, Shi X, Cai M, Cao C, Hu Z, Liu H, Tian J. Radiopharmaceuticals and Fluorescein Sodium Mediated Triple-Modality Molecular Imaging Allows Precise Image-Guided Tumor Surgery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900159. [PMID: 31380183 PMCID: PMC6662088 DOI: 10.1002/advs.201900159] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/01/2019] [Indexed: 05/06/2023]
Abstract
Radical resection is the most effective method for malignant tumor treatments. However, conventional imaging cannot fully satisfy the clinical needs of surgical navigation. This study presents a novel triple-modality positron emission tomography (PET)-Cerenkov radiation energy transfer (CRET)-confocal laser endomicroscopy (CLE) imaging strategy for intraoperative tumor imaging and surgical navigation. Using clinical radiopharmaceuticals and fluorescein sodium (FS), this strategy can accurately detect the tumor and guide the tumor surgery. The FS emission property under Cerenkov radiation excitation is investigated using 2-deoxy-2-18F-fluoroglucose and 11C-choline. Performances of the PET-CRET-CLE imaging and the CRET-CLE image-guided surgery are evaluated on mouse models. The CRET signal at 8 mm depth is stronger than the Cerenkov luminescence at 1 mm depth in phantoms. In vivo experiments indicate that 0.5 mL kg-1 of 10% FS generates the strongest CRET signal, which can be observed immediately after FS injection. A surgical navigation study shows that the tumors are precisely detected and resected using intraoperative CRET-CLE. In summary, a PET-CRET-CLE triple-modality imaging strategy is developed. This strategy can detect the tumors and precisely guide the tumor resection using clinical pharmaceuticals. This triple-modality imaging shows high potential in surgical navigation research and clinical translation.
Collapse
Affiliation(s)
- Sheng Zheng
- Department of GastroenterologyThe Third Medical CentreChinese PLA General HospitalBeijing100039China
- Department of GastroenterologyAnhui No.2 Provincial People's HospitalHefei230041China
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingThe State Key Laboratory of Management and Control for Complex SystemsInstitute of AutomationChinese Academy of SciencesUniversity of Chinese Academy of SciencesBeijing100190China
| | - Zeyu Zhang
- School of Life Science and TechnologyXidian UniversityXi'an710071China
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingThe State Key Laboratory of Management and Control for Complex SystemsInstitute of AutomationChinese Academy of SciencesUniversity of Chinese Academy of SciencesBeijing100190China
| | - Yawei Qu
- Department of GastroenterologyThe Third Medical CentreChinese PLA General HospitalBeijing100039China
| | - Xiaojun Zhang
- Department of Nuclear MedicineChinese PLA General HospitalBeijing100853China
| | - Hongbo Guo
- School of Information Sciences and TechnologyNorthwest UniversityXi'an710127China
| | - Xiaojing Shi
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingThe State Key Laboratory of Management and Control for Complex SystemsInstitute of AutomationChinese Academy of SciencesUniversity of Chinese Academy of SciencesBeijing100190China
| | - Meishan Cai
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingThe State Key Laboratory of Management and Control for Complex SystemsInstitute of AutomationChinese Academy of SciencesUniversity of Chinese Academy of SciencesBeijing100190China
| | - Caiguang Cao
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingThe State Key Laboratory of Management and Control for Complex SystemsInstitute of AutomationChinese Academy of SciencesUniversity of Chinese Academy of SciencesBeijing100190China
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingThe State Key Laboratory of Management and Control for Complex SystemsInstitute of AutomationChinese Academy of SciencesUniversity of Chinese Academy of SciencesBeijing100190China
| | - Haifeng Liu
- Department of GastroenterologyThe Third Medical CentreChinese PLA General HospitalBeijing100039China
| | - Jie Tian
- School of Life Science and TechnologyXidian UniversityXi'an710071China
- CAS Key Laboratory of Molecular ImagingBeijing Key Laboratory of Molecular ImagingThe State Key Laboratory of Management and Control for Complex SystemsInstitute of AutomationChinese Academy of SciencesUniversity of Chinese Academy of SciencesBeijing100190China
| |
Collapse
|
62
|
Waterhouse DJ, Fitzpatrick CRM, Pogue BW, O'Connor JPB, Bohndiek SE. A roadmap for the clinical implementation of optical-imaging biomarkers. Nat Biomed Eng 2019; 3:339-353. [PMID: 31036890 DOI: 10.1038/s41551-019-0392-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/17/2019] [Indexed: 02/07/2023]
Abstract
Clinical workflows for the non-invasive detection and characterization of disease states could benefit from optical-imaging biomarkers. In this Perspective, we discuss opportunities and challenges towards the clinical implementation of optical-imaging biomarkers for the early detection of cancer by analysing two case studies: the assessment of skin lesions in primary care, and the surveillance of patients with Barrett's oesophagus in specialist care. We stress the importance of technical and biological validations and clinical-utility assessments, and the need to address implementation bottlenecks. In addition, we define a translational roadmap for the widespread clinical implementation of optical-imaging technologies.
Collapse
Affiliation(s)
- Dale J Waterhouse
- Department of Physics, University of Cambridge, Cambridge, UK
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Catherine R M Fitzpatrick
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Engineering, University of Cambridge, Cambridge, UK
| | | | | | - Sarah E Bohndiek
- Department of Physics, University of Cambridge, Cambridge, UK.
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
63
|
Linssen MD, ter Weele EJ, Allersma DP, Lub-de Hooge MN, van Dam GM, Jorritsma-Smit A, Nagengast WB. Roadmap for the Development and Clinical Translation of Optical Tracers Cetuximab-800CW and Trastuzumab-800CW. J Nucl Med 2019; 60:418-423. [DOI: 10.2967/jnumed.118.216556] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/06/2018] [Indexed: 01/01/2023] Open
|
64
|
Stammes MA, Bugby SL, Porta T, Pierzchalski K, Devling T, Otto C, Dijkstra J, Vahrmeijer AL, de Geus-Oei LF, Mieog JSD. Modalities for image- and molecular-guided cancer surgery. Br J Surg 2018; 105:e69-e83. [PMID: 29341161 DOI: 10.1002/bjs.10789] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 10/25/2017] [Accepted: 11/05/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Surgery is the cornerstone of treatment for many solid tumours. A wide variety of imaging modalities are available before surgery for staging, although surgeons still rely primarily on visual and haptic cues in the operating environment. Image and molecular guidance might improve the adequacy of resection through enhanced tumour definition and detection of aberrant deposits. Intraoperative modalities available for image- and molecular-guided cancer surgery are reviewed here. METHODS Intraoperative cancer detection techniques were identified through a systematic literature search, with selection of peer-reviewed publications from January 2012 to January 2017. Modalities were reviewed, described and compared according to 25 predefined characteristics. To summarize the data in a comparable way, a three-point rating scale was applied to quantitative characteristics. RESULTS The search identified ten image- and molecular-guided surgery techniques, which can be divided into four groups: conventional, optical, nuclear and endogenous reflectance modalities. Conventional techniques are the most well known imaging modalities, but unfortunately have the drawback of a defined resolution and long acquisition time. Optical imaging is a real-time modality; however, the penetration depth is limited. Nuclear modalities have excellent penetration depth, but their intraoperative use is limited by the use of radioactivity. Endogenous reflectance modalities provide high resolution, although with a narrow field of view. CONCLUSION Each modality has its strengths and weaknesses; no single technique will be suitable for all surgical procedures. Strict selection of modalities per cancer type and surgical requirements is required as well as combining techniques to find the optimal balance.
Collapse
Affiliation(s)
- M A Stammes
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands.,Percuros, Enschede, The Netherlands
| | - S L Bugby
- Space Research Centre, Department of Physics and Astronomy, University of Leicester, Leicester, UK
| | - T Porta
- Maastricht MultiModal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| | - K Pierzchalski
- Maastricht MultiModal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, The Netherlands
| | | | - C Otto
- Medical Cell Bio Physics, University of Twente, Enschede, The Netherlands
| | - J Dijkstra
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Centre, Leiden, The Netherlands
| | - L-F de Geus-Oei
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands.,Biomedical Photonic Imaging Group, MIRA Institute, University of Twente, Enschede, The Netherlands
| | - J S D Mieog
- Department of Surgery, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
65
|
Feroldi F, Verlaan M, Knaus H, Davidoiu V, Vugts DJ, van Dongen GAMS, Molthoff CFM, de Boer JF. High resolution combined molecular and structural optical imaging of colorectal cancer in a xenograft mouse model. BIOMEDICAL OPTICS EXPRESS 2018; 9:6186-6204. [PMID: 31065422 PMCID: PMC6491025 DOI: 10.1364/boe.9.006186] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/01/2018] [Accepted: 11/06/2018] [Indexed: 05/08/2023]
Abstract
With the emergence of immunotherapies for cancer treatment, there is a rising clinical need to visualize the tumor microenvironment (TME) non-invasively in detail, which could be crucial to predict the efficacy of therapy. Nuclear imaging techniques enable whole-body imaging but lack the required spatial resolution. Conversely, near-infrared immunofluorescence (immuno-NIRF) is able to reveal tumor cells and/or other cell subsets in the TME by targeting the expression of a specific membrane receptor with fluorescently labeled monoclonal antibodies (mAb). Optical coherence tomography (OCT) provides three-dimensional morphological imaging of tissues without exogenous contrast agents. The combination of the two allows molecular and structural contrast at a resolution of ~15 µm, allowing for the specific location of a cell-type target with immuno-NIRF as well as revealing the three-dimensional architectural context with OCT. For the first time, combined immuno-NIRF and OCT of a tumor is demonstrated in situ in a xenograft mouse model of human colorectal cancer, targeted by a clinically-safe fluorescent mAb, revealing unprecedented details of the TME. A handheld scanner for ex vivo examination and an endoscope designed for imaging bronchioles in vivo are presented. This technique promises to complement nuclear imaging for diagnosing cancer invasiveness, precisely determining tumor margins, and studying the biodistribution of newly developed antibodies in high detail.
Collapse
Affiliation(s)
- Fabio Feroldi
- Department of Physics and Astronomy, LaserLaB Amsterdam, VU University Amsterdam, de Boelelaan 1081, 1081HV, Amsterdam, The Netherlands
| | - Mariska Verlaan
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Helene Knaus
- Department of Physics and Astronomy, LaserLaB Amsterdam, VU University Amsterdam, de Boelelaan 1081, 1081HV, Amsterdam, The Netherlands
| | - Valentina Davidoiu
- Department of Physics and Astronomy, LaserLaB Amsterdam, VU University Amsterdam, de Boelelaan 1081, 1081HV, Amsterdam, The Netherlands
| | - Danielle J. Vugts
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Guus A. M. S. van Dongen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Carla F. M. Molthoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Johannes F. de Boer
- Department of Physics and Astronomy, LaserLaB Amsterdam, VU University Amsterdam, de Boelelaan 1081, 1081HV, Amsterdam, The Netherlands
| |
Collapse
|
66
|
Meershoek P, van den Berg NS, Brouwer OR, Teertstra HJ, Lange CAH, Valdés-Olmos RA, van der Hiel B, Balm AJM, Klop WMC, van Leeuwen FWB. Three-Dimensional Tumor Margin Demarcation Using the Hybrid Tracer Indocyanine Green- 99mTc-Nanocolloid: A Proof-of-Concept Study in Tongue Cancer Patients Scheduled for Sentinel Node Biopsy. J Nucl Med 2018; 60:764-769. [PMID: 30504140 DOI: 10.2967/jnumed.118.220202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/15/2022] Open
Abstract
For radical resection of squamous cell carcinoma of the oral cavity, a tumor-free margin of at least 5 mm is required. Unfortunately, establishing in-depth margins is a surgical conundrum. Knowing that the hybrid sentinel node (SN) tracer indocyanine green (ICG)-99mTc-nanocolloid generates temporary tattoolike markings at the site of administration, we studied the ability to apply this tracer for tumor margin demarcation combined with SN biopsy. Methods: Nineteen patients with clinical T1-T2 oral tongue tumors received the traditional superficial 3 or 4 deposits of ICG-99mTc-nanocolloid (0.1 mL each), and in 12 patients additional deposits were placed deeply using ultrasound guidance (total of 6; 0.07 mL each). SN mapping was performed using lymphoscintigraphy and SPECT/CT. Before and directly after tumor excision, fluorescence imaging was performed to monitor the tracer deposits in the patient (fluorescent deposits were not used to guide the surgical excision). At pathologic examination, primary tumor samples were studied in detail. Results: The number of tracer depositions did not induce a significant difference in the number of SNs visualized (P = 0.836). Reproducible and deep tracer deposition proved to be challenging. The fluorescent nature of ICG-99mTc-nanocolloid supported in vivo and ex vivo identification of the tracer deposits surrounding the tumor. Pathologic examination indicated that in 66.7% (8/12), all fluorescence was observed within the resection margins. Conclusion: This study indicates that tumor margin demarcation combined with SN identification has potential but that some practical challenges need to be overcome if this technique is to mature as a surgical guidance concept. Future studies need to define whether the technology can improve the radical nature of the resections.
Collapse
Affiliation(s)
- Philippa Meershoek
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Head-and-Neck Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Nynke S van den Berg
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Oscar R Brouwer
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Jelle Teertstra
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| | - Charlotte A H Lange
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| | - Renato A Valdés-Olmos
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bernies van der Hiel
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Alfons J M Balm
- Department of Head-and-Neck Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - W Martin C Klop
- Department of Head-and-Neck Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands .,Department of Head-and-Neck Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| |
Collapse
|
67
|
Newton AD, Predina JD, Corbett CJ, Frenzel-Sulyok LG, Xia L, Petersson EJ, Tsourkas A, Nie S, Delikatny EJ, Singhal S. Optimization of Second Window Indocyanine Green for Intraoperative Near-Infrared Imaging of Thoracic Malignancy. J Am Coll Surg 2018; 228:188-197. [PMID: 30471345 DOI: 10.1016/j.jamcollsurg.2018.11.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Near-infrared (NIR) imaging using the second time window of indocyanine green (ICG) allows localization of pulmonary, pleural, and mediastinal malignancies during surgery. Based on empirical evidence, we hypothesized that different histologic tumor types fluoresce optimally at different ICG doses. STUDY DESIGN Patients with thoracic tumors biopsy-proven or suspicious for malignancy were enrolled in an NIR imaging clinical trial. Patients received a range of ICG doses 1 day before surgery: 1 mg/kg (n = 8), 2 mg/kg (n = 8), 3 mg/kg (n = 13), 4 mg/kg (n = 8), and 5 mg/kg (n = 8). Intraoperatively, NIR imaging was performed. The endpoint was to identify the highest tumor-to-background fluorescence ratio (TBR) for each tumor type at each dose. Final pathology confirmed tumor histology. RESULTS Of 45 patients, 41 had malignancies (18 non-small cell lung cancers [NSCLC], 3 pulmonary neuroendocrine tumors, 13 thoracic metastases, 4 thymomas, 3 mesotheliomas). At doses of 4 to 5 mg/kg, the TBR from primary NSCLC vs other malignancies was no different (2.70 vs 3.21, p = 1.00). At doses of 1 to 3 mg/kg, the TBR was greater for the NSCLCs (3.19 vs 1.49, p = 0.0006). Background fluorescence from the heart or ribs was observed in 1 of 16 cases at 1 to 2 mg/kg, 5 of 13 cases at 3 mg/kg, and 14 of 16 cases at 4 to 5 mg/kg; this was a major determinant of dose optimization. CONCLUSIONS This is the first study to demonstrate that the optimal NIR contrast agent dose varies by tumor histology. Lower dose ICG (2 to 3 mg/kg) is superior for nonprimary lung cancers, and high dose ICG (4 to 5 mg/kg) is superior for lung cancers. This will have major implications as more intraoperative imaging trials surface in other specialties, will significantly reduce costs and may facilitate wider application.
Collapse
Affiliation(s)
- Andrew D Newton
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.
| | - Jarrod D Predina
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Christopher J Corbett
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Lydia G Frenzel-Sulyok
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Leilei Xia
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - E James Petersson
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Shuming Nie
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Edward J Delikatny
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
68
|
Pogue BW, Rosenthal EL, Achilefu S, van Dam GM. Perspective review of what is needed for molecular-specific fluorescence-guided surgery. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-9. [PMID: 30291698 PMCID: PMC6210787 DOI: 10.1117/1.jbo.23.10.100601] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/07/2018] [Indexed: 05/21/2023]
Abstract
Molecular image-guided surgery has the potential for translating the tools of molecular pathology to real-time guidance in surgery. As a whole, there are incredibly positive indicators of growth, including the first United States Food and Drug Administration clearance of an enzyme-biosynthetic-activated probe for surgery guidance, and a growing number of companies producing agents and imaging systems. The strengths and opportunities must be continued but are hampered by important weaknesses and threats within the field. A key issue to solve is the inability of macroscopic imaging tools to resolve microscopic biological disease heterogeneity and the limitations in microscopic systems matching surgery workflow. A related issue is that parsing out true molecular-specific uptake from simple-enhanced permeability and retention is hard and requires extensive pathologic analysis or multiple in vivo tests, comparing fluorescence accumulation with standard histopathology and immunohistochemistry. A related concern in the field is the over-reliance on a finite number of chosen preclinical models, leading to early clinical translation when the probe might not be optimized for high intertumor variation or intratumor heterogeneity. The ultimate potential may require multiple probes, as are used in molecular pathology, and a combination with ultrahigh-resolution imaging and image recognition systems, which capture the data at a finer granularity than is possible by the surgeon. Alternatively, one might choose a more generalized approach by developing the tracer based on generic hallmarks of cancer to create a more "one-size-fits-all" concept, similar to metabolic aberrations as exploited in fluorodeoxyglucose - positron emission tomography (FDG-PET) (i.e., Warburg effect) or tumor acidity. Finally, methods to approach the problem of production cost minimization and regulatory approvals in a manner consistent with the potential revenue of the field will be important. In this area, some solid steps have been demonstrated in the use of fluorescent labeling commercial antibodies and separately in microdosing studies with small molecules.
Collapse
Affiliation(s)
- Brian W. Pogue
- Dartmouth College, Thayer School of Engineering and Department of Surgery, Geisel School of Medicine, Hanover, New Hampshire, United States
- Address all correspondence to: Brian W. Pogue, E-mail:
| | - Eben L. Rosenthal
- Stanford University School of Medicine, Department of Otolaryngology and Head and Neck Surgery, Stanford, California, United States
| | - Samuel Achilefu
- Washington University in St. Louis, Department of Radiology, St. Louis, Missouri, United States
| | - Gooitzen M. van Dam
- University Medical Center Groningen, Department of Surgery, Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
| |
Collapse
|
69
|
Favril S, Abma E, Blasi F, Stock E, Devriendt N, Vanderperren K, de Rooster H. Clinical use of organic near-infrared fluorescent contrast agents in image-guided oncologic procedures and its potential in veterinary oncology. Vet Rec 2018; 183:354. [DOI: 10.1136/vr.104851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/11/2018] [Accepted: 04/17/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Sophie Favril
- Small Animal Department, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| | - Eline Abma
- Small Animal Department, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| | - Francesco Blasi
- Ephoran Multi-Imaging Solutions s.r.l.; Colleretto Giacosa Italy
| | - Emmelie Stock
- Department of Medical Imaging of Domestic Animals, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Nausikaa Devriendt
- Small Animal Department, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Katrien Vanderperren
- Department of Medical Imaging of Domestic Animals, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Hilde de Rooster
- Small Animal Department, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| |
Collapse
|
70
|
Newton AD, Predina JD, Nie S, Low PS, Singhal S. Intraoperative fluorescence imaging in thoracic surgery. J Surg Oncol 2018; 118:344-355. [PMID: 30098293 DOI: 10.1002/jso.25149] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/04/2018] [Indexed: 12/19/2022]
Abstract
Intraoperative fluorescence imaging (IFI) can improve real-time identification of cancer cells during an operation. Phase I clinical trials in thoracic surgery have demonstrated that IFI with second window indocyanine green (TumorGlow® ) can identify subcentimeter pulmonary nodules, anterior mediastinal masses, and mesothelioma, while the use of a folate receptor-targeted near-infrared agent, OTL38, can improve the specificity for diagnosing tumors with folate receptor expression. Here, we review the existing preclinical and clinical data on IFI in thoracic surgery.
Collapse
Affiliation(s)
- Andrew D Newton
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jarrod D Predina
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shuming Nie
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
71
|
Lwin TM, Hoffman RM, Bouvet M. Advantages of patient-derived orthotopic mouse models and genetic reporters for developing fluorescence-guided surgery. J Surg Oncol 2018; 118:253-264. [PMID: 30080930 PMCID: PMC6146062 DOI: 10.1002/jso.25150] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022]
Abstract
Fluorescence-guided surgery can enhance the surgeon's ability to achieve a complete oncologic resection. There are a number of tumor-specific probes being developed with many preclinical mouse models to evaluate their efficacy. The current review discusses the different preclinical mouse models in the setting of probe evaluation and highlights the advantages of patient-derived orthotopic xenografts (PDOX) mouse models and genetic reporters to develop fluorescence-guided surgery.
Collapse
Affiliation(s)
- Thinzar M. Lwin
- Department of Surgery, University of California San Diego, San Diego, CA
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, San Diego, CA
- AntiCancer, Inc., San Diego, CA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, CA
- Department of Surgery, VA Medical Center, San Diego, CA
| |
Collapse
|
72
|
Hentzen JE, de Jongh SJ, Hemmer PH, van der Plas WY, van Dam GM, Kruijff S. Molecular fluorescence-guided surgery of peritoneal carcinomatosis of colorectal origin: A narrative review. J Surg Oncol 2018; 118:332-343. [PMID: 29938400 PMCID: PMC6174973 DOI: 10.1002/jso.25106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/16/2018] [Accepted: 04/22/2018] [Indexed: 12/14/2022]
Abstract
Patients with peritoneal carcinomatosis (PC) from colorectal origin may undergo cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) as a curative approach. One major prognostic factor that affects survival is completeness of cytoreduction. Molecular Fluorescence Guided Surgery (MFGS) is a novel intraoperative imaging technique that may improve tumor identification in the future, potentially preventing over- and under-treatment in these patients. This narrative review outlines a chronological overview of MFGS development in patients with PC of colorectal origin.
Collapse
Affiliation(s)
- Judith E.K.R. Hentzen
- Department of Surgery, Division of Surgical Oncology, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Steven J. de Jongh
- Department of Gastroenterology and Hepatology, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Patrick H.J. Hemmer
- Department of Surgery, Division of Surgical Oncology, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Willemijn Y. van der Plas
- Department of Surgery, Division of Surgical Oncology, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| | - Gooitzen M. van Dam
- Department of Surgery, Division of Surgical Oncology, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Nuclear Medicine and Molecular Imaging and Intensive Care, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Schelto Kruijff
- Department of Surgery, Division of Surgical Oncology, University Medical Centre GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
73
|
Tummers WS, Willmann JK, Bonsing BA, Vahrmeijer AL, Gambhir SS, Swijnenburg RJ. Advances in Diagnostic and Intraoperative Molecular Imaging of Pancreatic Cancer. Pancreas 2018; 47:675-689. [PMID: 29894417 PMCID: PMC6003672 DOI: 10.1097/mpa.0000000000001075] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis. To improve outcomes, there is a critical need for improved tools for detection, accurate staging, and resectability assessment. This could improve patient stratification for the most optimal primary treatment modality. Molecular imaging, used in combination with tumor-specific imaging agents, can improve established imaging methods for PDAC. These novel, tumor-specific imaging agents developed to target specific biomarkers have the potential to specifically differentiate between malignant and benign diseases, such as pancreatitis. When these agents are coupled to various types of labels, this type of molecular imaging can provide integrated diagnostic, noninvasive imaging of PDAC as well as image-guided pancreatic surgery. This review provides a detailed overview of the current clinical imaging applications, upcoming molecular imaging strategies for PDAC, and potential targets for imaging, with an emphasis on intraoperative imaging applications.
Collapse
Affiliation(s)
- Willemieke S. Tummers
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA. Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Juergen K. Willmann
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA. Juergen K. Willmann died January 8, 2018
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sanjiv S. Gambhir
- Address correspondence to: R.J. Swijnenburg, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands (). Tel: +31 71 526 4005, Fax: +31 71 526 6750
| | - Rutger-Jan Swijnenburg
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
74
|
Wu C, Gleysteen J, Teraphongphom NT, Li Y, Rosenthal E. In-vivo optical imaging in head and neck oncology: basic principles, clinical applications and future directions. Int J Oral Sci 2018; 10:10. [PMID: 29555901 PMCID: PMC5944254 DOI: 10.1038/s41368-018-0011-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 12/29/2017] [Accepted: 01/10/2018] [Indexed: 02/05/2023] Open
Abstract
Head and neck cancers become a severe threat to human's health nowadays and represent the sixth most common cancer worldwide. Surgery remains the first-line choice for head and neck cancer patients. Limited resectable tissue mass and complicated anatomy structures in the head and neck region put the surgeons in a dilemma between the extensive resection and a better quality of life for the patients. Early diagnosis and treatment of the pre-malignancies, as well as real-time in vivo detection of surgical margins during en bloc resection, could be leveraged to minimize the resection of normal tissues. With the understanding of the head and neck oncology, recent advances in optical hardware and reagents have provided unique opportunities for real-time pre-malignancies and cancer imaging in the clinic or operating room. Optical imaging in the head and neck has been reported using autofluorescence imaging, targeted fluorescence imaging, high-resolution microendoscopy, narrow band imaging and the Raman spectroscopy. In this study, we reviewed the basic theories and clinical applications of optical imaging for the diagnosis and treatment in the field of head and neck oncology with the goal of identifying limitations and facilitating future advancements in the field.
Collapse
Affiliation(s)
- Chenzhou Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - John Gleysteen
- Department of Otolaryngology, University of Tennessee Health Science Center, 38163, Memphis, TN, USA
| | | | - Yi Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Eben Rosenthal
- Department of Otolaryngology and Radiology, Stanford University, 94305, Stanford, CA, USA.
| |
Collapse
|
75
|
Vidal Fortuny J, Sadowski SM, Belfontali V, Guigard S, Poncet A, Ris F, Karenovics W, Triponez F. Randomized clinical trial of intraoperative parathyroid gland angiography with indocyanine green fluorescence predicting parathyroid function after thyroid surgery. Br J Surg 2018; 105:350-357. [PMID: 29405252 PMCID: PMC6084300 DOI: 10.1002/bjs.10783] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/15/2017] [Accepted: 11/08/2017] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hypoparathyroidism, the most common complication after thyroid surgery, leads to hypocalcaemia and significant medical problems. An RCT was undertaken to determine whether intraoperative parathyroid gland angiography with indocyanine green (ICG) could predict postoperative hypoparathyroidism, and obviate the need for systematic blood tests and oral calcium supplementation. METHODS Between September 2014 and February 2016, patients who had at least one well perfused parathyroid gland on ICG angiography were randomized to receive standard follow-up (measurement of calcium and parathyroid hormone (PTH) on postoperative day (POD) 1 and systematic supplementation with calcium and vitamin D; control group) or no supplementation and no blood test on POD 1 (intervention group). In all patients, calcium and PTH levels were measured 10-15 days after thyroidectomy. The primary endpoint was hypocalcaemia on POD 10-15. RESULTS A total of 196 patients underwent ICG angiography during thyroid surgery, of whom 146 had at least one well perfused parathyroid gland on ICG angiography and were randomized. None of these patients presented with hypoparathyroidism, including those who did not receive calcium supplementation. The intervention group was statistically non-inferior to the control group (exact 95 per cent c.i. of the difference in proportion of patients with hypocalcaemia -0·053 to 0·053; P = 0·012). Eleven of the 50 excluded patients, in whom no well perfused parathyroid gland could be identified by angiography, presented with hypoparathyroidism on POD 1, and six on POD 10-15, which was significantly different from the findings in randomized patients (P = 0·007). CONCLUSION ICG angiography reliably predicts the vascularization of the parathyroid glands and obviates the need for postoperative measurement of calcium and PTH, and supplementation with calcium in patients with at least one well perfused parathyroid gland. Registration number: NCT02249780 (http://www.clinicaltrials.gov).
Collapse
Affiliation(s)
- J. Vidal Fortuny
- Department of Thoracic and Endocrine Surgery and Faculty of MedicineUniversity Hospitals of GenevaGenevaSwitzerland
| | - S. M. Sadowski
- Department of Thoracic and Endocrine Surgery and Faculty of MedicineUniversity Hospitals of GenevaGenevaSwitzerland
| | - V. Belfontali
- Department of Thoracic and Endocrine Surgery and Faculty of MedicineUniversity Hospitals of GenevaGenevaSwitzerland
| | - S. Guigard
- Department of Thoracic and Endocrine Surgery and Faculty of MedicineUniversity Hospitals of GenevaGenevaSwitzerland
| | - A. Poncet
- Department of Clinical EpidemiologyUniversity Hospitals of GenevaGenevaSwitzerland
| | - F. Ris
- Department Visceral SurgeryUniversity Hospitals of GenevaGenevaSwitzerland
| | - W. Karenovics
- Department of Thoracic and Endocrine Surgery and Faculty of MedicineUniversity Hospitals of GenevaGenevaSwitzerland
| | - F. Triponez
- Department of Thoracic and Endocrine Surgery and Faculty of MedicineUniversity Hospitals of GenevaGenevaSwitzerland
| |
Collapse
|
76
|
Qiu SQ, Zhang GJ, Jansen L, de Vries J, Schröder CP, de Vries EGE, van Dam GM. Evolution in sentinel lymph node biopsy in breast cancer. Crit Rev Oncol Hematol 2018; 123:83-94. [PMID: 29482783 DOI: 10.1016/j.critrevonc.2017.09.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/12/2017] [Accepted: 09/19/2017] [Indexed: 02/05/2023] Open
Abstract
Sentinel lymph node biopsy (SLNB) is the standard of care for axillary staging in clinically node-negative (cN0) breast cancer patients without neoadjuvant chemotherapy (NAC). The application of SLNB in patients receiving NAC has also been explored. Evidence supports its use after NAC in pretreatment cN0 patients. Nonetheless, its routine use in all the pretreatment node-positive patients who become cN0 after NAC is unjustified due to the unacceptably high false-negative rate, which can be improved in a subset of patients. Axillary surgery omission in selected patients with a low risk of ALN metastasis has gained more and more research interest because the SLNs are tumor-free in more than 70% of all patients. To avoid drawbacks of conventional mapping methods, novel techniques for SLN detection have been developed and shown to be highly accurate in patients with early breast cancer. This article reviews the progress in SLNB in patients with breast cancer.
Collapse
Affiliation(s)
- Si-Qi Qiu
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands; Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands; The Breast Center, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Guo-Jun Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Guangdong, China; Changjiang Scholar's Laboratory of Shantou University Medical College, Guangdong, China
| | - Liesbeth Jansen
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Jakob de Vries
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolien P Schröder
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Gooitzen M van Dam
- Department of Surgery, University Medical Center Groningen, Groningen, The Netherlands; Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
77
|
Wilhelm D, Vogel T, Ostler D, Marahrens N, Kohn N, Koller S, Friess H, Kranzfelder M. Enhanced Visualization: From Intraoperative Tissue Differentiation to Augmented Reality. Visc Med 2018; 34:52-59. [PMID: 29594170 DOI: 10.1159/000485940] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Optimal visualization of the operative field and methods that additionally provide supportive optical information form the basis for target-directed and successful surgery. This article strives to give an overview of current enhanced visualization techniques in visceral surgery and to highlight future developments. Methods The article was written as a comprehensive review on this topic and is based on a MEDLINE search and ongoing research from our own group and from other working groups. Results Various techniques for enhanced visualization are described comprising augmented reality, unspecific and targeted staining methods, and optical modalities such as narrow-band imaging. All facilitate our surgical performance; however, due to missing randomized controlled studies for most of the innovations reported on, the available evidence is low. Conclusion Many new visualization technologies are emerging with the aim to improve our perception of the surgical field leading to less invasive, target-oriented, and elegant treatment forms that are of significant benefit to our patients.
Collapse
Affiliation(s)
- Dirk Wilhelm
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Working Group for Minimally Invasive Interdisciplinary Therapeutical Intervention, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Vogel
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Working Group for Minimally Invasive Interdisciplinary Therapeutical Intervention, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Daniel Ostler
- Working Group for Minimally Invasive Interdisciplinary Therapeutical Intervention, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nils Marahrens
- Working Group for Minimally Invasive Interdisciplinary Therapeutical Intervention, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nils Kohn
- Working Group for Minimally Invasive Interdisciplinary Therapeutical Intervention, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sebastian Koller
- Working Group for Minimally Invasive Interdisciplinary Therapeutical Intervention, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael Kranzfelder
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Working Group for Minimally Invasive Interdisciplinary Therapeutical Intervention, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
78
|
Hartshorn CM, Bradbury MS, Lanza GM, Nel AE, Rao J, Wang AZ, Wiesner UB, Yang L, Grodzinski P. Nanotechnology Strategies To Advance Outcomes in Clinical Cancer Care. ACS NANO 2018; 12:24-43. [PMID: 29257865 PMCID: PMC6589353 DOI: 10.1021/acsnano.7b05108] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Ongoing research into the application of nanotechnology for cancer treatment and diagnosis has demonstrated its advantages within contemporary oncology as well as its intrinsic limitations. The National Cancer Institute publishes the Cancer Nanotechnology Plan every 5 years since 2005. The most recent iteration helped codify the ongoing basic and translational efforts of the field and displayed its breadth with several evolving areas. From merely a technological perspective, this field has seen tremendous growth and success. However, an incomplete understanding of human cancer biology persists relative to the application of nanoscale materials within contemporary oncology. As such, this review presents several evolving areas in cancer nanotechnology in order to identify key clinical and biological challenges that need to be addressed to improve patient outcomes. From this clinical perspective, a sampling of the nano-enabled solutions attempting to overcome barriers faced by traditional therapeutics and diagnostics in the clinical setting are discussed. Finally, a strategic outlook of the future is discussed to highlight the need for next-generation cancer nanotechnology tools designed to address critical gaps in clinical cancer care.
Collapse
Affiliation(s)
- Christopher M Hartshorn
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Author,
| | - Michelle S Bradbury
- Department of Radiology and Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, New York, 10065, United States
| | - Gregory M Lanza
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63108, United States
| | - Andre E Nel
- Division of NanoMedicine, Department of Medicine, and California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jianghong Rao
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, California 94305, United States
| | - Andrew Z. Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Ulrich B Wiesner
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14843, United States
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Piotr Grodzinski
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Author,
| |
Collapse
|
79
|
Abstract
Progressive technological advancements in imaging have significantly improved the preoperative sensitivity for the detection of very small foci of regionally- or hematogenously-metastatic colorectal cancer. Unfortunately, this information has not translated to continued linear gains in patient survival, and might even result in the false-positive upstaging of some cases: these are two conundrums in the imaging of colorectal cancer. Both conundrums might be resolved by the widespread use of real-time imaging guidance during operative procedures. This might open the way for the widespread use of fluorodeoxyglucose PET/CT for the initial staging of patients with colorectal cancer.
Collapse
Affiliation(s)
- Nathan C Hall
- Department of Radiology, Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; Diagnostic Imaging, Nuclear Medicine, Corporal Michael J. Crescenz VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA 19104, USA; Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 410 West 10th Avenue, Columbus, OH 43210, USA.
| | - Alexander T Ruutiainen
- Diagnostic Radiology, Corporal Michael J. Crescenz VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA 19104, USA
| |
Collapse
|
80
|
Napp J, Stammes MA, Claussen J, Prevoo HA, Sier CF, Hoeben FJ, Robillard MS, Vahrmeijer AL, Devling T, Chan AB, de Geus-Oei LF, Alves F. Fluorescence- and multispectral optoacoustic imaging for an optimized detection of deeply located tumors in an orthotopic mouse model of pancreatic carcinoma. Int J Cancer 2018; 142:2118-2129. [DOI: 10.1002/ijc.31236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/04/2017] [Accepted: 12/14/2017] [Indexed: 01/30/2023]
Affiliation(s)
- Joanna Napp
- Institute of Interventional and Diagnostic Radiology, University Medical Center Göttingen; Göttingen Lower Saxony Germany
- Clinic of Haematology and Medical Oncology; University Medical Center Göttingen; Göttingen Lower Saxony Germany
- Translational Molecular Imaging, Max-Planck-Institute of Experimental Medicine; Göttingen Lower Saxony Germany
| | - Marieke A. Stammes
- Percuros B.V., AE Enschede; The Netherlands
- Department of Radiology; Leiden University Medical Center; RC Leiden The Netherlands
| | - Jing Claussen
- iThera Medical GmbH, Zielstattstrasse; Munich Germany
| | | | | | | | - Marc S. Robillard
- Tagworks Pharmaceuticals, Geert Grooteplein Zuid 10; GA Nijmegen The Netherlands
| | | | - Tim Devling
- iThera Medical GmbH, Zielstattstrasse; Munich Germany
| | | | - Lioe-Fee de Geus-Oei
- Department of Radiology; Leiden University Medical Center; RC Leiden The Netherlands
- Biomedical Photonic Imaging Group, MIRA Institute, University of Twente; AE Enschede The Netherlands
| | - Frauke Alves
- Institute of Interventional and Diagnostic Radiology, University Medical Center Göttingen; Göttingen Lower Saxony Germany
- Clinic of Haematology and Medical Oncology; University Medical Center Göttingen; Göttingen Lower Saxony Germany
- Translational Molecular Imaging, Max-Planck-Institute of Experimental Medicine; Göttingen Lower Saxony Germany
| |
Collapse
|
81
|
Nagaya T, Nakamura YA, Choyke PL, Kobayashi H. Fluorescence-Guided Surgery. Front Oncol 2017; 7:314. [PMID: 29312886 PMCID: PMC5743791 DOI: 10.3389/fonc.2017.00314] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/05/2017] [Indexed: 01/02/2023] Open
Abstract
Surgical resection of cancer remains an important treatment modality. Despite advances in preoperative imaging, surgery itself is primarily guided by the surgeon’s ability to locate pathology with conventional white light imaging. Fluorescence-guided surgery (FGS) can be used to define tumor location and margins during the procedure. Intraoperative visualization of tumors may not only allow more complete resections but also improve safety by avoiding unnecessary damage to normal tissue which can also reduce operative time and decrease the need for second-look surgeries. A number of new FGS imaging probes have recently been developed, complementing a small but useful number of existing probes. In this review, we describe current and new fluorescent probes that may assist FGS.
Collapse
Affiliation(s)
- Tadanobu Nagaya
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yu A Nakamura
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
82
|
Sadowski SM, Vidal Fortuny J, Triponez F. A reappraisal of vascular anatomy of the parathyroid gland based on fluorescence techniques. Gland Surg 2017; 6:S30-S37. [PMID: 29322020 DOI: 10.21037/gs.2017.07.10] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Identification of the parathyroid glands (PGs) during thyroid surgery may prevent their inadvertent surgical removal and prevent postoperative hypoparathyroidism. However, identification of the PGs does not guarantee their function, and their vascular supply needs to be preserved as well. The recent introduction of intraoperative indocyanine green (ICG) fluorescent angiography of the PGs during thyroid surgery allows for the appraisal of the vascular anatomy and evaluation of PG function. The use of this tool could lead to a significant reduction in the rate of postoperative hypoparathyroidism, as it allows surgeons to adapt their surgical technique for the preservation of the PGs. ICG fluorescent angiography is currently the only available real-time tool to assess the vascular blood supply of each individual PG intraoperatively and can thus assist surgeons in their decision-making. Herein, we review the relevant literature.
Collapse
Affiliation(s)
| | - Jordi Vidal Fortuny
- Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
| | - Frederic Triponez
- Thoracic and Endocrine Surgery, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
83
|
Upchurch E, Griffiths S, Lloyd GR, Isabelle M, Kendall C, Barr H. Developments in optical imaging for gastrointestinal surgery. Future Oncol 2017; 13:2363-2382. [PMID: 29121775 DOI: 10.2217/fon-2017-0181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
To improve outcomes for patients with cancer, in terms of both survival and a reduction in the morbidity and mortality that results from surgical resection and treatment, there are two main areas that require improvement. Accurate early diagnosis of the cancer, at a stage where curative and, ideally, minimally invasive treatment is achievable, is desired as well as identification of tumor margins, lymphatic and distant disease, enabling complete, but not unnecessarily extensive, resection. Optical imaging is making progress in achieving these aims. This review discusses the principles of optical imaging, focusing on fluorescence and spectroscopy, and the current research that is underway in GI tract carcinomas.
Collapse
Affiliation(s)
- Emma Upchurch
- Biophotonics Research Unit, Gloucestershire Royal Hospital, Great Western Road, Gloucester, UK, GL1 3NN.,Department of Upper GI Surgery, Gloucestershire Royal Hospital, Great Western Road, Gloucester, UK, GL1 3NN
| | - Shelly Griffiths
- Department of Upper GI Surgery, Gloucestershire Royal Hospital, Great Western Road, Gloucester, UK, GL1 3NN
| | - Gavin-Rhys Lloyd
- Biophotonics Research Unit, Gloucestershire Royal Hospital, Great Western Road, Gloucester, UK, GL1 3NN
| | - Martin Isabelle
- Renishaw plc, New Mills, Wotton-under-Edge, Gloucestershire, UK, GL12 8JR
| | - Catherine Kendall
- Biophotonics Research Unit, Gloucestershire Royal Hospital, Great Western Road, Gloucester, UK, GL1 3NN
| | - Hugh Barr
- Biophotonics Research Unit, Gloucestershire Royal Hospital, Great Western Road, Gloucester, UK, GL1 3NN.,Department of Upper GI Surgery, Gloucestershire Royal Hospital, Great Western Road, Gloucester, UK, GL1 3NN
| |
Collapse
|
84
|
De Leeuw F, Breuskin I, Abbaci M, Casiraghi O, Mirghani H, Ben Lakhdar A, Laplace-Builhé C, Hartl D. Intraoperative Near-infrared Imaging for Parathyroid Gland Identification by Auto-fluorescence: A Feasibility Study. World J Surg 2017; 40:2131-8. [PMID: 27220510 DOI: 10.1007/s00268-016-3571-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Parathyroid glands (PGs) can be particularly hard to distinguish from surrounding tissue and thus can be damaged or removed during thyroidectomy. Postoperative hypoparathyroidism is the most common complication after thyroidectomy. Very recently, it has been found that the parathyroid tissue shows near-infrared (NIR) auto-fluorescence which could be used for intraoperative detection, without any use of contrast agents. The work described here presents a histological validation ex vivo of the NIR imaging procedure and evaluates intraoperative PG detection by NIR auto-fluorescence using for the first time to our knowledge a commercially available clinical NIR imaging device. METHODS Ex vivo study on resected operative specimens combined with a prospective in vivo study of consecutive patients who underwent total or partial thyroid, or parathyroid surgery at a comprehensive cancer center. During surgery, any tissue suspected to be a potential PG by the surgeon was imaged with the Fluobeam 800 (®) system. NIR imaging was compared to conventional histology (ex vivo) and/or visual identification by the surgeon (in vivo). RESULTS We have validated NIR auto-fluorescence with an ex vivo study including 28 specimens. Sensitivity and specificity were 94.1 and 80 %, respectively. Intraoperative NIR imaging was performed in 35 patients and 81 parathyroids were identified. In 80/81 cases, the fluorescence signal was subjectively obvious on real-time visualization. We determined that PG fluorescence is 2.93 ± 1.59 times greater than thyroid fluorescence in vivo. CONCLUSIONS Real-time NIR imaging based on parathyroid auto-fluorescence is fast, safe, and non-invasive and shows very encouraging results, for intraoperative parathyroid identification.
Collapse
Affiliation(s)
- Frederic De Leeuw
- Plateforme d'Imagerie et Cytométrie, UMS AMMICa, Gustave Roussy, Université Paris-Saclay, 114, rue Édouard-Vaillant, 94805, Villejuif, France.
- UMR CNRS 8081- IR4M, Université Paris-Sud, Université Paris-Saclay, 91401, Orsay, France.
| | - Ingrid Breuskin
- Département de Chirurgie ORL, Unité Thyroïde, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Muriel Abbaci
- Plateforme d'Imagerie et Cytométrie, UMS AMMICa, Gustave Roussy, Université Paris-Saclay, 114, rue Édouard-Vaillant, 94805, Villejuif, France
- UMR CNRS 8081- IR4M, Université Paris-Sud, Université Paris-Saclay, 91401, Orsay, France
| | - Odile Casiraghi
- Département de Pathologie, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Haïtham Mirghani
- Département de Chirurgie ORL, Unité Thyroïde, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Aïcha Ben Lakhdar
- Département de Pathologie, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| | - Corinne Laplace-Builhé
- Plateforme d'Imagerie et Cytométrie, UMS AMMICa, Gustave Roussy, Université Paris-Saclay, 114, rue Édouard-Vaillant, 94805, Villejuif, France
- UMR CNRS 8081- IR4M, Université Paris-Sud, Université Paris-Saclay, 91401, Orsay, France
| | - Dana Hartl
- Département de Chirurgie ORL, Unité Thyroïde, Gustave Roussy, Université Paris-Saclay, 94805, Villejuif, France
| |
Collapse
|
85
|
Jitpratoom P, Anuwong A. The use of ICG enhanced fluorescence for the evaluation of parathyroid gland preservation. Gland Surg 2017; 6:579-586. [PMID: 29142851 DOI: 10.21037/gs.2017.09.01] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Indocyanine green (ICG) enhanced fluorescence imaging is recent innovation as the "real-time intraoperative imaging" technique. Many clinical studies have been reported in the literature which use different devices and techniques that employ various doses and usages of ICG as a non-specific contrast agent. Several groups have performed studies in endocrine surgery, especially with regards to parathyroid-related outcomes after thyroid and parathyroid surgery. However, there is no consensus on the technical details that should be applied. With this study, we aimed to review the current literature on potential use of intraoperative ICG angiography for evaluating parathyroid gland (PTG) preservation.
Collapse
Affiliation(s)
| | - Angkoon Anuwong
- Department of Surgery, Police General Hospital, Bangkok, Thailand
| |
Collapse
|
86
|
Jones A, Wilton J. Can intra-operative fluorescence play a significant role in hepatobiliary surgery? Eur J Surg Oncol 2017; 43:1622-1627. [DOI: 10.1016/j.ejso.2017.02.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/17/2017] [Accepted: 02/24/2017] [Indexed: 01/02/2023] Open
|
87
|
Garcia M, Gruev V. Optical characterization of rigid endoscopes and polarization calibration methods. OPTICS EXPRESS 2017; 25:15713-15728. [PMID: 28789084 DOI: 10.1364/oe.25.015713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/20/2017] [Indexed: 06/07/2023]
Abstract
Polarization imaging can reveal orthogonal information with respect to color about the structural composition of biological tissue, and with the advance of superior polarimeters its use for biomedical applications has proliferated in the last decade. Polarimetry can be used in pre-clinical and clinical settings for the early detection of cancerous tissue. Polarization-based endoscopy with the complementary near-infrared fluorescence imaging modality improves the early diagnosis of flat cancerous lesions in colorectal tumor models. With the development of new polarization sensors the need to use standard laboratory optics to create custom imaging systems increases. These additional optics can behave as polarization filters effectively degrading and modifying the original tissue's polarization signatures leading to erroneous judgments. Here, we present a framework to characterize the spectral and polarization properties of rigid endoscopes for polarization-based endoscopic imaging. We describe and evaluate two calibration schemes based on Mueller calculus to reconstruct the original polarization information. Optical limitations of the endoscopes and minimum polarimeter requirements are discussed that may be of interest to other researchers working with custom polarization-based imaging systems.
Collapse
|
88
|
Abstract
The unique spectral signatures and biologically inert compositions of surface-enhanced resonance Raman scattering (SERRS) nanoparticles make them promising contrast agents for in vivo cancer imaging. Our SERRS nanoparticles consist of a 60-nm gold nanoparticle core that is encapsulated in a 15-nm-thick silica shell wherein the resonant Raman reporter is embedded. Subtle aspects of their preparation can shift their limit of detection by orders of magnitude. In this protocol, we present the optimized, step-by-step procedure for generating reproducible SERRS nanoparticles with femtomolar (10-15 M) limits of detection. We provide ways of characterizing the optical properties of SERRS nanoparticles using UV/VIS and Raman spectroscopy, and their physicochemical properties using transmission electron microscopy and nanoparticle tracking analysis. We introduce several applications of these nanoprobes for biomedical research, with a focus on intraoperative cancer imaging via Raman imaging. A detailed account is provided for successful i.v. administration of SERRS nanoparticles such that delineation of cancerous lesions can be achieved in vivo and ex vivo on resected tissues without the need for specific biomarker targeting. This straightforward, yet comprehensive, protocol-from initial de novo gold nanoparticle synthesis to SERRS nanoparticle contrast-enhanced preclinical Raman imaging in animal models-takes ∼96 h.
Collapse
|
89
|
The promising impact of molecular profiling on treatment strategies in oral cancers. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2017; 118:242-247. [PMID: 28576460 DOI: 10.1016/j.jormas.2017.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/22/2017] [Indexed: 12/19/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is a major cause of cancer-associated morbidity and mortality. Although OSCC may develop from easily accessible oral preneoplastic lesions (OPLs), no intervention has been reported so far that reduces the rate of malignant transformation. A comprehensive molecular characterization of oral carcinogenesis may help refining treatment strategies both in patients with OPLs and OSCC. Herein, we review main molecular alterations occurring at different steps during oral carcinogenesis and show how molecularly-based medicine and surgery may impact the outcome of OSCC in the future.
Collapse
|
90
|
Rungsakulkij N, Tangtawee P. Fluorescence cholangiography during laparoscopic cholecystectomy in a patient with situs inversus totalis: a case report and literature review. BMC Surg 2017; 17:43. [PMID: 28427402 PMCID: PMC5397776 DOI: 10.1186/s12893-017-0242-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/11/2017] [Indexed: 12/17/2022] Open
Abstract
Background Situs inversus totalis is a rare autosomal disorder in which the patient’s affected visceral organs are a perfect mirror image of their normal positions. Surgery in these patients is technically challenging. Minimally invasive surgery such as laparoscopic cholecystectomy is the standard treatment for symptomatic cholelithiasis, but it can be difficult to perform. Laparoscopic cholecystectomy in patients with situs inversus totalis may be even more technically challenging. Fluorescence cholangiography is a new innovation in the field of navigation surgery. This procedure is safe and easy to perform, its findings are easy to interpret, and it does not require a learning curve or radiographs. It can be used in real time during surgery to identify extrahepatic biliary structures. Case presentation We herein report a case of situs inversus totalis in a Thai patient with a history of biliary pancreatitis. He underwent laparoscopic cholecystectomy with intraoperative fluorescence cholangiography. The operation was successfully completed without complications. To the best of our knowledge, this is the first case report of the use of fluorescence cholangiography during laparoscopic cholecystectomy in a patient with situs inversus. Conclusion Fluorescence cholangiographyis a new navigational surgical technique with which to identify extrahepatic biliary structures. It can be used as an adjunct technique during laparoscopic cholecystectomy to avoid biliary tract injury in difficult cases. Electronic supplementary material The online version of this article (doi:10.1186/s12893-017-0242-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Narongsak Rungsakulkij
- Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand.
| | - Pongsatorn Tangtawee
- Department of Surgery, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
91
|
Pogue BW, Paulsen KD, Samkoe KS, Elliott JT, Hasan T, Strong TV, Draney DR, Feldwisch J. Vision 20/20: Molecular-guided surgical oncology based upon tumor metabolism or immunologic phenotype: Technological pathways for point of care imaging and intervention. Med Phys 2017; 43:3143-3156. [PMID: 27277060 DOI: 10.1118/1.4951732] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Surgical guidance with fluorescence has been demonstrated in individual clinical trials for decades, but the scientific and commercial conditions exist today for a dramatic increase in clinical value. In the past decade, increased use of indocyanine green based visualization of vascular flow, biliary function, and tissue perfusion has spawned a robust growth in commercial systems that have near-infrared emission imaging and video display capabilities. This recent history combined with major preclinical innovations in fluorescent-labeled molecular probes, has the potential for a shift in surgical practice toward resection guidance based upon molecular information in addition to conventional visual and palpable cues. Most surgical subspecialties already have treatment management decisions partially based upon the immunohistochemical phenotype of the cancer, as assessed from molecular pathology of the biopsy tissue. This phenotyping can inform the surgical resection process by spatial mapping of these features. Further integration of the diagnostic and therapeutic value of tumor metabolism sensing molecules or immune binding agents directly into the surgical process can help this field mature. Maximal value to the patient would come from identifying the spatial patterns of molecular expression in vivo that are well known to exist. However, as each molecular agent is advanced into trials, the performance of the imaging system can have a critical impact on the success. For example, use of pre-existing commercial imaging systems are not well suited to image receptor targeted fluorophores because of the lower concentrations expected, requiring orders of magnitude more sensitivity. Additionally the imaging system needs the appropriate dynamic range and image processing features to view molecular probes or therapeutics that may have nonspecific uptake or pharmacokinetic issues which lead to limitations in contrast. Imaging systems need to be chosen based upon objective performance criteria, and issues around calibration, validation, and interpretation need to be established before a clinical trial starts. Finally, as early phase trials become more established, the costs associated with failures can be crippling to the field, and so judicious use of phase 0 trials with microdose levels of agents is one viable paradigm to help the field advance, but this places high sensitivity requirements on the imaging systems used. Molecular-guided surgery has truly transformative potential, and several key challenges are outlined here with the goal of seeing efficient advancement with ideal choices. The focus of this vision 20/20 paper is on the technological aspects that are needed to be paired with these agents.
Collapse
Affiliation(s)
- Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755 and Department of Surgery, Dartmouth College, Hanover, New Hampshire 03755
| | - Keith D Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755; Department of Surgery, Dartmouth College, Hanover, New Hampshire 03755; and Department of Diagnostic Radiology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire 03755
| | - Kimberley S Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755 and Department of Surgery, Dartmouth College, Hanover, New Hampshire 03755
| | - Jonathan T Elliott
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114 and Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Theresa V Strong
- Vector Production Facility, Division of Hematology Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | | | | |
Collapse
|
92
|
Chromophores in operative surgery: Current practice and rationalized development. J Control Release 2017; 249:123-130. [DOI: 10.1016/j.jconrel.2016.12.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/23/2016] [Accepted: 12/28/2016] [Indexed: 12/18/2022]
|
93
|
Image-guided surgery: Transistor-like pH nanoprobes. Nat Biomed Eng 2017. [DOI: 10.1038/s41551-016-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
94
|
Lamberts LE, Koch M, de Jong JS, Adams ALL, Glatz J, Kranendonk MEG, Terwisscha van Scheltinga AGT, Jansen L, de Vries J, Lub-de Hooge MN, Schröder CP, Jorritsma-Smit A, Linssen MD, de Boer E, van der Vegt B, Nagengast WB, Elias SG, Oliveira S, Witkamp AJ, Mali WPTM, Van der Wall E, van Diest PJ, de Vries EGE, Ntziachristos V, van Dam GM. Tumor-Specific Uptake of Fluorescent Bevacizumab-IRDye800CW Microdosing in Patients with Primary Breast Cancer: A Phase I Feasibility Study. Clin Cancer Res 2016; 23:2730-2741. [PMID: 28119364 DOI: 10.1158/1078-0432.ccr-16-0437] [Citation(s) in RCA: 199] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 10/21/2016] [Accepted: 10/22/2016] [Indexed: 12/22/2022]
Abstract
Purpose: To provide proof of principle of safety, breast tumor-specific uptake, and positive tumor margin assessment of the systemically administered near-infrared fluorescent tracer bevacizumab-IRDye800CW targeting VEGF-A in patients with breast cancer.Experimental Design: Twenty patients with primary invasive breast cancer eligible for primary surgery received 4.5 mg bevacizumab-IRDye800CW as intravenous bolus injection. Safety aspects were assessed as well as tracer uptake and tumor delineation during surgery and ex vivo in surgical specimens using an optical imaging system. Ex vivo multiplexed histopathology analyses were performed for evaluation of biodistribution of tracer uptake and coregistration of tumor tissue and healthy tissue.Results: None of the patients experienced adverse events. Tracer levels in primary tumor tissue were higher compared with those in the tumor margin (P < 0.05) and healthy tissue (P < 0.0001). VEGF-A tumor levels also correlated with tracer levels (r = 0.63, P < 0.0002). All but one tumor showed specific tracer uptake. Two of 20 surgically excised lumps contained microscopic positive margins detected ex vivo by fluorescent macro- and microscopy and confirmed at the cellular level.Conclusions: Our study shows that systemic administration of the bevacizumab-IRDye800CW tracer is safe for breast cancer guidance and confirms tumor and tumor margin uptake as evaluated by a systematic validation methodology. The findings are a step toward a phase II dose-finding study aimed at in vivo margin assessment and point to a novel drug assessment tool that provides a detailed picture of drug distribution in the tumor tissue. Clin Cancer Res; 23(11); 2730-41. ©2016 AACR.
Collapse
Affiliation(s)
- Laetitia E Lamberts
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Maximillian Koch
- Technische Universität München & Helmholtz Zentrum, München, Germany
| | - Johannes S de Jong
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Arthur L L Adams
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jürgen Glatz
- Technische Universität München & Helmholtz Zentrum, München, Germany
| | - Mariëtte E G Kranendonk
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Anton G T Terwisscha van Scheltinga
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Hospital and Clinical Pharmacy, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Liesbeth Jansen
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jakob de Vries
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marjolijn N Lub-de Hooge
- Hospital and Clinical Pharmacy, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Carolien P Schröder
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Annelies Jorritsma-Smit
- Hospital and Clinical Pharmacy, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Matthijs D Linssen
- Hospital and Clinical Pharmacy, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Esther de Boer
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bert van der Vegt
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wouter B Nagengast
- Department of Gastroenterology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sjoerd G Elias
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sabrina Oliveira
- Division of Cell Biology of the Department of Biology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Arjen J Witkamp
- Department of Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Willem P Th M Mali
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Elsken Van der Wall
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Gooitzen M van Dam
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Intensive Care, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
95
|
[Intraoperative multidimensional visualization]. Chirurg 2016; 87:1015-1024. [PMID: 27796416 DOI: 10.1007/s00104-016-0314-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Modern intraoperative techniques of visualization are increasingly being applied in general and visceral surgery. The combination of diverse techniques provides the possibility of multidimensional intraoperative visualization of specific anatomical structures. Thus, it is possible to differentiate between normal tissue and tumor tissue and therefore exactly define tumor margins. The aim of intraoperative visualization of tissue that is to be resected and tissue that should be spared is to lead to a rational balance between oncological and functional results. Moreover, these techniques help to analyze the physiology and integrity of tissues. Using these methods surgeons are able to analyze tissue perfusion and oxygenation. However, to date it is not clear to what extent these imaging techniques are relevant in the clinical routine. The present manuscript reviews the relevant modern visualization techniques focusing on intraoperative computed tomography and magnetic resonance imaging as well as augmented reality, fluorescence imaging and optoacoustic imaging.
Collapse
|
96
|
Smolensky D, Rathore K, Cekanova M. Molecular targets in urothelial cancer: detection, treatment, and animal models of bladder cancer. Drug Des Devel Ther 2016; 10:3305-3322. [PMID: 27784990 PMCID: PMC5063594 DOI: 10.2147/dddt.s112113] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bladder cancer remains one of the most expensive cancers to treat in the United States due to the length of required treatment and degree of recurrence. In order to treat bladder cancer more effectively, targeted therapies are being investigated. In order to use targeted therapy in a patient, it is important to provide a genetic background of the patient. Recent advances in genome sequencing, as well as transcriptome analysis, have identified major pathway components altered in bladder cancer. The purpose of this review is to provide a broad background on bladder cancer, including its causes, diagnosis, stages, treatments, animal models, as well as signaling pathways in bladder cancer. The major focus is given to the PI3K/AKT pathway, p53/pRb signaling pathways, and the histone modification machinery. Because several promising immunological therapies are also emerging in the treatment of bladder cancer, focus is also given on general activation of the immune system for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Dmitriy Smolensky
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine
- UT-ORNL Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN, USA
| | - Kusum Rathore
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine
| | - Maria Cekanova
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine
- UT-ORNL Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
97
|
Winum JY. Synthesis and composition of amino acid linking groups conjugated to compounds used for the targeted imaging of tumors: a patent evaluation of US20160011199A1. Expert Opin Ther Pat 2016; 26:1223-1226. [DOI: 10.1080/13543776.2016.1241238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Jean-Yves Winum
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université de Montpellier, ENSCM, Ecole Nationale Supérieure de Chimie de Montpellier, Montpellier Cedex, France
| |
Collapse
|
98
|
Vidal Fortuny J, Karenovics W, Triponez F, Sadowski SM. Intra-Operative Indocyanine Green Angiography of the Parathyroid Gland. World J Surg 2016; 40:2378-81. [PMID: 26944954 PMCID: PMC5028398 DOI: 10.1007/s00268-016-3493-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Major complications of thyroid and parathyroid surgery are recurrent laryngeal nerve injuries and definitive hypoparathyroidism. The use of intra-operative Indocyanine Green Angiography for confirmation of vascular status of the parathyroid gland is reported here.
Collapse
Affiliation(s)
- Jordi Vidal Fortuny
- Thoracic and Endocrine Surgery, University Hospitals of Geneva, 1211, Geneva, Switzerland
| | - Wolfram Karenovics
- Thoracic and Endocrine Surgery, University Hospitals of Geneva, 1211, Geneva, Switzerland
| | - Frederic Triponez
- Thoracic and Endocrine Surgery, University Hospitals of Geneva, 1211, Geneva, Switzerland
| | - Samira M Sadowski
- Thoracic and Endocrine Surgery, University Hospitals of Geneva, 1211, Geneva, Switzerland.
| |
Collapse
|
99
|
Harlaar NJ, Koller M, de Jongh SJ, van Leeuwen BL, Hemmer PH, Kruijff S, van Ginkel RJ, Been LB, de Jong JS, Kats-Ugurlu G, Linssen MD, Jorritsma-Smit A, van Oosten M, Nagengast WB, Ntziachristos V, van Dam GM. Molecular fluorescence-guided surgery of peritoneal carcinomatosis of colorectal origin: a single-centre feasibility study. Lancet Gastroenterol Hepatol 2016; 1:283-290. [PMID: 28404198 DOI: 10.1016/s2468-1253(16)30082-6] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Optimum cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is essential for the curative treatment of peritoneal carcinomatosis of colorectal origin. At present, surgeons depend on visual inspection and palpation for tumour detection. Improved detection of tumour tissue using molecular fluorescence-guided surgery could not only help attain a complete cytoreduction of metastatic lesions, but might also prevent overtreatment by avoiding resection of benign lesions. METHODS For this non-randomised, single-centre feasibility study, we enrolled patients with colorectal peritoneal metastases scheduled for cytoreductive surgery and HIPEC. 2 days before surgery, 4·5 mg of the near-infrared fluorescent tracer bevacizumab-IRDye800CW was administered intravenously. The primary objectives were to determine the safety and feasibility of molecular fluorescence-guided surgery using bevacizumab-IRDye800CW. Molecular fluorescence-guided surgery was deemed safe if no allergic or anaphylactic reactions were recorded and no serious adverse events were attributed to bevacizumab-IRDye800CW. The technique was deemed feasible if bevacizumab-IRDye800CW enabled detection of fluorescence signals intraoperatively. Secondary objectives were correlation of fluorescence with histopathology by back-table imaging of the fresh surgical specimen and semi-quantitative ex-vivo analyses of formalin-fixed paraffin embedded (FFPE) tissue on all peritoneal lesions. Additionally, VEGF-α staining and fluorescence microscopy was done. This study is registered with the Netherlands Trial Registry, number NTR4632. FINDINGS Between July 3, 2014, and March 2, 2015, seven patients were enrolled in the study. One patient developed an abdominal sepsis 5 days postoperatively and another died from an asystole 4 days postoperatively, most probably due to a cardiovascular thromboembolic event. However, both serious adverse events were attributed to the surgical cytoreductive surgery and HIPEC procedure. No serious adverse events related to bevacizumab-IRDye800CW occurred in any of the patients. Intraoperatively, fluorescence was seen in all patients. In two patients, additional tumour tissue was detected by molecular fluorescence-guided surgery that was initially missed by the surgeons. During back-table imaging of fresh surgical specimens, a total of 80 areas were imaged, marked, and analysed. All of the 29 non-fluorescent areas were found to contain only benign tissue, whereas tumour tissue was detected in 27 of 51 fluorescent areas (53%). Ex-vivo semi-quantification of 79 FFPE peritoneal lesions showed a tumour-to-normal ratio of 6·92 (SD 2·47). INTERPRETATION Molecular fluorescence-guided surgery using the near-infrared fluorescent tracer bevacizumab-IRDye800CW is safe and feasible. This technique might be of added value for the treatment of patients with colorectal peritoneal metastases through improved patient selection and optimisation of cytoreductive surgery. A subsequent multicentre phase 2 trial is needed to make a definitive assessment of the diagnostic accuracy and the effect on clinical decision making of molecular fluorescence-guided surgery. FUNDING FP-7 Framework Programme BetaCure and SurgVision BV.
Collapse
Affiliation(s)
- Niels J Harlaar
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjory Koller
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Steven J de Jongh
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Barbara L van Leeuwen
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Patrick H Hemmer
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Schelto Kruijff
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Robert J van Ginkel
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lukas B Been
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Johannes S de Jong
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands; Department of Pathology, University Medical Center of Utrecht, Utrecht, Netherlands
| | - Gursah Kats-Ugurlu
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Matthijs D Linssen
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands; Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Annelies Jorritsma-Smit
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marleen van Oosten
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wouter B Nagengast
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Vasilis Ntziachristos
- Technical University of Munich and Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Munich, Germany
| | - Gooitzen M van Dam
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands; Department of Nuclear Medicine and Molecular Imaging and Intensive Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
100
|
Indocyanine Green Angiography in Subtotal Parathyroidectomy: Technique for the Function of the Parathyroid Remnant. J Am Coll Surg 2016; 223:e43-e49. [PMID: 27568330 DOI: 10.1016/j.jamcollsurg.2016.08.540] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 08/16/2016] [Indexed: 11/21/2022]
|