1
|
Karsalia R, Zhou CC, Muhammad N, Teng CW, Singh Y, Huang V, Harmsen S, Lee JYK. Dose optimization of second window indocyanine green in meningioma patients. Clin Neurol Neurosurg 2024; 243:108385. [PMID: 38878642 DOI: 10.1016/j.clineuro.2024.108385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/08/2024] [Indexed: 07/14/2024]
Abstract
OBJECTIVE Surgery remains the first line treatment for meningiomas and can benefit from fluorescence-guided surgical techniques such as second-window indocyanine green (SWIG). In the current study, we compared the use of the standard SWIG dose of 5.0 mg/kg relative to 2.5 mg/kg indocyanine green (ICG) in meningioma patients. METHODS Patients were prospectively enrolled in an IRB-approved study of SWIG and received either the standard dose of 5.0 mg/kg or a reduced dose of 2.5 mg/kg of ICG around 24 h prior to their surgery. Intraoperative near-infrared fluorescence imaging was performed with exo- and endoscopic systems. Signal-to-background ratio (SBR) was calculated to quantify fluorescence and was compared between 5.0 mg/kg and 2.5 mg/kg ICG. All patients received pre-operative MRI and, in select cases, the pre-operative MRI was correlated to intraoperative fluorescence imaging. RESULTS/DISCUSSION In the current study, we found no significant difference in the SBR of meningiomas in patients that were administered with either 5.0 mg/kg or 2.5 mg/kg ICG. However, in five patients that received the standard-dose SWIG regimen of 5.0 mg/kg ICG we observed dose-related fluorescence quenching - referred to as "inversion" - that interfered with tumor visualization during fluorescence-guided surgery (FGS). When correlated to pre-operative MRI, a similar rim pattern was observed around the primary tumor on T2 FLAIR, which, in retrospect, could be used as a predictor for inversion during FGS in meningioma patients receiving standard-dose ICG. CONCLUSION This study demonstrated that a reduced ICG dose was as effective as standard-dose SWIG in meningioma patients. We therefore recommend to adjust the standard ICG dose for meningioma patients to 2.5 mg/kg particularly when rim enhancement is observed on pre-operative T2 FLAIR.
Collapse
Affiliation(s)
- Ritesh Karsalia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Cecilia C Zhou
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Najib Muhammad
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Clare W Teng
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yash Singh
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Vincent Huang
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Stefan Harmsen
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Picart T, Gautheron A, Caredda C, Ray C, Mahieu-Williame L, Montcel B, Guyotat J. Fluorescence-Guided Surgical Techniques in Adult Diffuse Low-Grade Gliomas: State-of-the-Art and Emerging Techniques: A Systematic Review. Cancers (Basel) 2024; 16:2698. [PMID: 39123426 PMCID: PMC11311317 DOI: 10.3390/cancers16152698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Diffuse low-grade gliomas are infiltrative tumors whose margins are not distinguishable from the adjacent healthy brain parenchyma. The aim was to precisely examine the results provided by the intraoperative use of macroscopic fluorescence in diffuse low-grade gliomas and to describe the new fluorescence-based techniques capable of guiding the resection of low-grade gliomas. Only about 20% and 50% of low-grade gliomas are macroscopically fluorescent after 5-amino-levulinic acid (5-ALA) or fluorescein sodium intake, respectively. However, 5-ALA is helpful for detecting anaplastic foci, and thus choosing the best biopsy targets in diffuse gliomas. Spectroscopic detection of 5-ALA-induced fluorescence can detect very low and non-macroscopically visible concentrations of protoporphyrin IX, a 5-ALA metabolite, and, consequently, has excellent performances for the detection of low-grade gliomas. Moreover, these tumors have a specific spectroscopic signature with two fluorescence emission peaks, which is useful for distinguishing them not only from healthy brain but also from high-grade gliomas. Confocal laser endomicroscopy can generate intraoperative optic biopsies, but its sensitivity remains limited. In the future, the coupled measurement of autofluorescence and induced fluorescence, and the introduction of fluorescence detection technologies providing a wider field of view could result in the development of operator-friendly tools implementable in the operative routine.
Collapse
Affiliation(s)
- Thiebaud Picart
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Groupe Hospitalier Est, Hospices Civils de Lyon, 59 Boulevard Pinel, 69500 Bron, France
- Faculty of Medicine Lyon Est, Université Claude Bernard Lyon 1, 8 Avenue Rockefeller, 69003 Lyon, France
- Cancer Research Centre of Lyon (CRCL) Inserm 1052, CNRS 5286, 28 Rue Laennec, 69008 Lyon, France
| | - Arthur Gautheron
- Laboratoire Hubert Curien UMR 5516, Institut d’Optique Graduate School, CNRS, Université Jean Monnet Saint-Etienne, 42023 Saint-Etienne, France;
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Charly Caredda
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Cédric Ray
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Laurent Mahieu-Williame
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Bruno Montcel
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| | - Jacques Guyotat
- Department of Neurosurgery, Hôpital Neurologique Pierre Wertheimer, Groupe Hospitalier Est, Hospices Civils de Lyon, 59 Boulevard Pinel, 69500 Bron, France
- Faculty of Medicine Lyon Est, Université Claude Bernard Lyon 1, 8 Avenue Rockefeller, 69003 Lyon, France
- CREATIS CNRS, Inserm, UMR 5220, U1294, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, 69100 Lyon, France; (C.C.); (C.R.); (L.M.-W.); (B.M.)
| |
Collapse
|
3
|
Radtke K, Schulz-Schaeffer WJ, Oertel J. Confocal laser endomicroscopy in glial tumors-a histomorphological analysis. Neurosurg Rev 2024; 47:65. [PMID: 38265724 PMCID: PMC10808457 DOI: 10.1007/s10143-024-02286-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 01/25/2024]
Abstract
OBJECTIVE The extent of resection and neurological outcome are important prognostic markers for overall survival in glioma patients. Confocal laser endomicroscopy is a tool to examine tissue without the need for fixation or staining. This study aims to analyze gliomas in confocal laser endomicroscopy and identify reliable diagnostic criteria for glial matter and glial tumors. MATERIAL AND METHODS One-hundred-and-five glioma specimens were analyzed using a 670-nm confocal laser endomicroscope and then processed into hematoxylin-eosin-stained frozen sections. All confocal images and frozen sections were evaluated for the following criteria: presence of tumor, cellularity, nuclear pleomorphism, changes of the extracellular glial matrix, microvascular proliferation, necrosis, and mitotic activity. Recurring characteristics were identified. Accuracy, sensitivity, specificity, and positive and negative predictive values were assessed for each feature. RESULTS All 125 specimens could be processed and successfully analyzed via confocal laser endomicroscopy. We found diagnostic criteria to identify white and grey matter and analyze cellularity, nuclear pleomorphism, changes in the glial matrix, vascularization, and necrosis in glial tumors. An accuracy of > 90.0 % was reached for grey matter, cellularity, and necrosis, > 80.0 % for white matter and nuclear pleomorphism, and > 70.0 % for microvascular proliferation and changes of the glial matrix. Mitotic activity could not be identified. Astroglial tumors showed significantly less nuclear pleomorphism in confocal laser endomicroscopy than oligodendroglial tumors (p < 0.001). Visualization of necrosis aids in the differentiation of low grade gliomas and high grade gliomas (p < 0.002). CONCLUSION Autofluorescence-based confocal laser endomicroscopy proved not only useful in differentiation between tumor and brain tissue but also revealed useful clues to further characterize tissue without processing in a lab. Possible applications include the improvement of extent of resection and the safe harvest of representative tissue for histopathological and molecular genetic diagnostics.
Collapse
Affiliation(s)
- Karen Radtke
- Klinik für Neurochirurgie, Medizinische Fakultät, Universität des Saarlandes, /Saar, 66421, Homburg, Germany
| | - Walter J Schulz-Schaeffer
- Institut für Neuropathologie, Medizinische Fakultät, Universität des Saarlandes, /Saar, 66421, Homburg, Germany
| | - Joachim Oertel
- Klinik für Neurochirurgie, Medizinische Fakultät, Universität des Saarlandes, /Saar, 66421, Homburg, Germany.
| |
Collapse
|
4
|
Bianconi A, Bonada M, Zeppa P, Colonna S, Tartara F, Melcarne A, Garbossa D, Cofano F. How Reliable Is Fluorescence-Guided Surgery in Low-Grade Gliomas? A Systematic Review Concerning Different Fluorophores. Cancers (Basel) 2023; 15:4130. [PMID: 37627158 PMCID: PMC10452554 DOI: 10.3390/cancers15164130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Fluorescence-guided surgery has been increasingly used to support glioma surgery with the purpose of obtaining a maximal safe resection, in particular in high-grade gliomas, while its role is less definitely assessed in low-grade gliomas. METHODS A systematic review was conducted. 5-aminolevulinic acid, sodium fluorescein, indocyanine green and tozuleristide were taken into account. The main considered outcome was the fluorescence rate, defined as the number of patients in whom positive fluorescence was detected out of the total number of patients. Only low-grade gliomas were considered, and data were grouped according to single fluorophores. RESULTS 16 papers about 5-aminolevulinic acid, 4 about sodium fluorescein, 2 about indocyanine green and 1 about tozuleristide were included in the systematic review. Regarding 5-aminolevulinic acid, a total of 467 low-grade glioma patients were included, and fluorescence positivity was detected in 34 out of 451 Grade II tumors (7.3%); while in Grade I tumors, fluorescence positivity was detected in 9 out of 16 cases. In 16 sodium fluorescein patients, seven positive fluorescent cases were detected. As far as indocyanine is concerned, two studies accounting for six patients (three positive) were included, while for tozuleristide, a single clinical trial with eight patients (two positive) was retrieved. CONCLUSIONS The current evidence does not support the routine use of 5-aminolevulinic acid or sodium fluorescein with a standard operating microscope because of the low fluorescence rates. New molecules, including tozuleristide, and new techniques for fluorescence detection have shown promising results; however, their use still needs to be clinically validated on a large scale.
Collapse
Affiliation(s)
- Andrea Bianconi
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Marta Bonada
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Pietro Zeppa
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Stefano Colonna
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Fulvio Tartara
- Headache Science and Neurorehabilitation Center, IRCCS Mondino Foundation, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Antonio Melcarne
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Diego Garbossa
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Fabio Cofano
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
- Humanitas Gradenigo, 10100 Turin, Italy
| |
Collapse
|
5
|
Polanski WH, Oelschlägel M, Juratli TA, Wahl H, Krukowski PM, Morgenstern U, Koch E, Steiner G, Schackert G, Sobottka SB. Topographic Mapping of the Primary Sensory Cortex Using Intraoperative Optical Imaging and Tactile Irritation. Brain Topogr 2023; 36:1-9. [PMID: 36446998 PMCID: PMC9834102 DOI: 10.1007/s10548-022-00925-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/30/2022] [Indexed: 11/30/2022]
Abstract
The determination of exact tumor boundaries within eloquent brain regions is essential to maximize the extent of resection. Recent studies showed that intraoperative optical imaging (IOI) combined with median nerve stimulation is a helpful tool for visualization of the primary sensory cortex (PSC). In this technical note, we describe a novel approach of using IOI with painless tactile irritation to demonstrate the feasibility of topographic mapping of different body regions within the PSC. In addition, we compared the IOI results with preoperative functional MRI (fMRI) findings. In five patients with tumors located near the PSC who received tumor removal, IOI with tactile irritation of different body parts and fMRI was applied. We showed that tactile irritation of the hand in local and general anesthesia leads to reliable changes of cerebral blood volume during IOI. Hereby, we observed comparable IOI activation maps regarding the median nerve stimulation, fMRI and tactile irritation of the hand. The tactile irritation of different body areas revealed a plausible topographic distribution along the PSC. With this approach, IOI is also suitable for awake surgeries, since the tactile irritation is painless compared with median nerve stimulation and is congruent to fMRI findings. Further studies are ongoing to standardize this method to enable a broad application within the neurosurgical community.
Collapse
Affiliation(s)
- Witold H. Polanski
- Department of Neurosurgery, University Hospital of Dresden, Fiedlerstr. 74, 01307 Dresden, Germany
| | - Martin Oelschlägel
- Clinical Sensoring and Monitoring, Faculty of Medicine, University Hospital Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Tareq A. Juratli
- Department of Neurosurgery, University Hospital of Dresden, Fiedlerstr. 74, 01307 Dresden, Germany
| | - Hannes Wahl
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Pawel M. Krukowski
- Institute of Diagnostic and Interventional Neuroradiology, University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Ute Morgenstern
- Institute of Biomedical Engineering, Faculty of Electrical and Computer Engineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Edmund Koch
- Clinical Sensoring and Monitoring, Faculty of Medicine, University Hospital Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Gerald Steiner
- Clinical Sensoring and Monitoring, Faculty of Medicine, University Hospital Carl Gustav Carus, Dresden University of Technology, 01307 Dresden, Germany
| | - Gabriele Schackert
- Department of Neurosurgery, University Hospital of Dresden, Fiedlerstr. 74, 01307 Dresden, Germany
| | - Stephan B. Sobottka
- Department of Neurosurgery, University Hospital of Dresden, Fiedlerstr. 74, 01307 Dresden, Germany
| |
Collapse
|
6
|
Schupper AJ, Rao M, Mohammadi N, Baron R, Lee JYK, Acerbi F, Hadjipanayis CG. Fluorescence-Guided Surgery: A Review on Timing and Use in Brain Tumor Surgery. Front Neurol 2021; 12:682151. [PMID: 34220688 PMCID: PMC8245059 DOI: 10.3389/fneur.2021.682151] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
Fluorescence-guided surgery (FGS) allows surgeons to have improved visualization of tumor tissue in the operating room, enabling maximal safe resection of malignant brain tumors. Over the past two decades, multiple fluorescent agents have been studied for FGS, including 5-aminolevulinic acid (5-ALA), fluorescein sodium, and indocyanine green (ICG). Both non-targeted and targeted fluorescent agents are currently being used in clinical practice, as well as under investigation, for glioma visualization and resection. While the efficacy of intraoperative fluorescence in studied fluorophores has been well established in the literature, the effect of timing on fluorophore administration in glioma surgery has not been as well depicted. In the past year, recent studies of 5-ALA use have shown that intraoperative fluorescence may persist beyond the previously studied window used in prior multicenter trials. Additionally, the use of fluorophores for different brain tumor types is discussed in detail, including a discussion of choosing the right fluorophore based on tumor etiology. In the following review, the authors will describe the temporal nature of the various fluorophores used in glioma surgery, what remains uncertain in FGS, and provide a guide for using fluorescence as a surgical adjunct in brain tumor surgery.
Collapse
Affiliation(s)
- Alexander J Schupper
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Manasa Rao
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nicki Mohammadi
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rebecca Baron
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - John Y K Lee
- Department of Neurosurgery, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Francesco Acerbi
- Department of Neurosurgery, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo Besta, Milan, Italy
| | | |
Collapse
|
7
|
Use of Second Window ICG in spinal cord biopsy of a mildly contrast-enhancing lesion: Technical note and review of the literature. Neurochirurgie 2021; 68:239-242. [PMID: 34102223 DOI: 10.1016/j.neuchi.2021.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Indocyanine green (ICG) is commonly used to visualize cerebral vasculature, particularly in the management of cerebral aneurysms. There have also been attempts to use ICG for visualization of tumors. Injection of ICG followed by immediate fluorescence microscopy is limited by the short time window for imaging and administration and restricted depth of imaging. Second Window Indocyanine Green (SWIG) addresses these issues by allowing for longer contrast times and the imaging of deeper regions of brain tissue. Biopsy of spinal cord lesions is often difficult for a variety of reasons, including the delicate nature of the tissue and differentiating normal from lesional tissue visually, especially in lesions with heterogeneous enhancement. METHODS In this case report, we describe the use of second window ICG to facilitate the visualization of a spinal cord lesion and subsequent biopsy of the lesion. RESULTS This patient is a 24-year-old female who had recurrence of a suprasellar germinoma. An MRI of the rest of the neuraxis was performed to assess for the presence of drop metastases. The spinal cord from C2-5 was expanded with areas of patchy enhancement; however, this lesion was asymptomatic. The patient's oncologist requested a biopsy of this lesion to help direct subsequent care of her recurrent germinoma. The day before surgery, the patient had an intravenous injection of ICG dye. She then underwent a C3-5 laminectomy for biopsy of her cervical intramedullary lesion. After opening of the dura, no visible abnormality of the spinal cord could be identified. A Stryker endoscope showed an area of ICG uptake in the cord at approximately the C3-4 level. A midline myelotomy was centered over the ICG demarcated area and several samples were taken for pathology. Final biopsy results determined the lesion to be spinal cord parenchyma with perivascular and intraparenchymal lymphocytes - not consistent with spinal cord tumor or germinoma. CONCLUSION Second Window ICG is effective in visualizing otherwise visually unremarkable spinal cord lesions. This technology can facilitate biopsy of these lesions and possibly their surgical resection.
Collapse
|
8
|
Schupper AJ, Yong RL, Hadjipanayis CG. The Neurosurgeon's Armamentarium for Gliomas: An Update on Intraoperative Technologies to Improve Extent of Resection. J Clin Med 2021; 10:jcm10020236. [PMID: 33440712 PMCID: PMC7826675 DOI: 10.3390/jcm10020236] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/18/2022] Open
Abstract
Maximal safe resection is the standard of care in the neurosurgical treatment of high-grade gliomas. To aid surgeons in the operating room, adjuvant techniques and technologies centered around improving intraoperative visualization of tumor tissue have been developed. In this review, we will discuss the most advanced technologies, specifically fluorescence-guided surgery, intraoperative imaging, neuromonitoring modalities, and microscopic imaging techniques. The goal of these technologies is to improve detection of tumor tissue beyond what conventional microsurgery has permitted. We describe the various advances, the current state of the literature that have tested the utility of the different adjuvants in clinical practice, and future directions for improving intraoperative technologies.
Collapse
|
9
|
Teng CW, Huang V, Arguelles GR, Zhou C, Cho SS, Harmsen S, Lee JYK. Applications of indocyanine green in brain tumor surgery: review of clinical evidence and emerging technologies. Neurosurg Focus 2021; 50:E4. [PMID: 33386005 DOI: 10.3171/2020.10.focus20782] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/23/2020] [Indexed: 11/06/2022]
Abstract
Indocyanine green (ICG) is a water-soluble dye that was approved by the FDA for biomedical purposes in 1956. Initially used to measure cardiocirculatory and hepatic functions, ICG's fluorescent properties in the near-infrared (NIR) spectrum soon led to its application in ophthalmic angiography. In the early 2000s, ICG was formally introduced in neurosurgery as an angiographic tool. In 2016, the authors' group pioneered a novel technique with ICG named second-window ICG (SWIG), which involves infusion of a high dose of ICG (5.0 mg/kg) in patients 24 hours prior to surgery. To date, applications of SWIG have been reported in patients with high-grade gliomas, meningiomas, brain metastases, pituitary adenomas, craniopharyngiomas, chordomas, and pinealomas.The applications of ICG have clearly expanded rapidly across different specialties since its initial development. As an NIR fluorophore, ICG has advantages over other FDA-approved fluorophores, all of which are currently in the visible-light spectrum, because of NIR fluorescence's increased tissue penetration and decreased autofluorescence. Recently, interest in the latest applications of ICG in brain tumor surgery has grown beyond its role as an NIR fluorophore, extending into shortwave infrared imaging and integration into nanotechnology. This review aims to summarize reported clinical studies on ICG fluorescence-guided surgery of intracranial tumors, as well as to provide an overview of the literature on emerging technologies related to the utility of ICG in neuro-oncological surgeries, including the following aspects: 1) ICG fluorescence in the NIR-II window; 2) ICG for photoacoustic imaging; and 3) ICG nanoparticles for combined diagnostic imaging and therapy (theranostic) applications.
Collapse
Affiliation(s)
- Clare W Teng
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and.,2Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vincent Huang
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and.,2Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gabriel R Arguelles
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and.,2Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cecilia Zhou
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and.,2Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steve S Cho
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and
| | - Stefan Harmsen
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and
| | - John Y K Lee
- 1Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia; and
| |
Collapse
|
10
|
Orillac C, Stummer W, Orringer DA. Fluorescence Guidance and Intraoperative Adjuvants to Maximize Extent of Resection. Neurosurgery 2020; 89:727-736. [PMID: 33289518 DOI: 10.1093/neuros/nyaa475] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 08/23/2020] [Indexed: 12/27/2022] Open
Abstract
Safely maximizing extent of resection has become the central goal in glioma surgery. Especially in eloquent cortex, the goal of maximal resection is balanced with neurological risk. As new technologies emerge in the field of neurosurgery, the standards for maximal safe resection have been elevated. Fluorescence-guided surgery, intraoperative magnetic resonance imaging, and microscopic imaging methods are among the most well-validated tools available to enhance the level of accuracy and safety in glioma surgery. Each technology uses a different characteristic of glioma tissue to identify and differentiate tumor tissue from normal brain and is most effective in the context of anatomic, connectomic, and neurophysiologic context. While each tool is able to enhance resection, multiple modalities are often used in conjunction to achieve maximal safe resection. This paper reviews the mechanism and utility of the major adjuncts available for use in glioma surgery, especially in tumors within eloquent areas, and puts forth the foundation for a unified approach to how leverage currently available technology to ensure maximal safe resection.
Collapse
Affiliation(s)
- Cordelia Orillac
- Department of Neurosurgery, NYU Langone Health, New York, New York
| | - Walter Stummer
- Department of Neurosurgery, University Hospital Münster, Münster, Germany
| | | |
Collapse
|
11
|
Belykh E, Shaffer KV, Lin C, Byvaltsev VA, Preul MC, Chen L. Blood-Brain Barrier, Blood-Brain Tumor Barrier, and Fluorescence-Guided Neurosurgical Oncology: Delivering Optical Labels to Brain Tumors. Front Oncol 2020; 10:739. [PMID: 32582530 PMCID: PMC7290051 DOI: 10.3389/fonc.2020.00739] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/17/2020] [Indexed: 12/17/2022] Open
Abstract
Recent advances in maximum safe glioma resection have included the introduction of a host of visualization techniques to complement intraoperative white-light imaging of tumors. However, barriers to the effective use of these techniques within the central nervous system remain. In the healthy brain, the blood-brain barrier ensures the stability of the sensitive internal environment of the brain by protecting the active functions of the central nervous system and preventing the invasion of microorganisms and toxins. Brain tumors, however, often cause degradation and dysfunction of this barrier, resulting in a heterogeneous increase in vascular permeability throughout the tumor mass and outside it. Thus, the characteristics of both the blood-brain and blood-brain tumor barriers hinder the vascular delivery of a variety of therapeutic substances to brain tumors. Recent developments in fluorescent visualization of brain tumors offer improvements in the extent of maximal safe resection, but many of these fluorescent agents must reach the tumor via the vasculature. As a result, these fluorescence-guided resection techniques are often limited by the extent of vascular permeability in tumor regions and by the failure to stain the full volume of tumor tissue. In this review, we describe the structure and function of both the blood-brain and blood-brain tumor barriers in the context of the current state of fluorescence-guided imaging of brain tumors. We discuss features of currently used techniques for fluorescence-guided brain tumor resection, with an emphasis on their interactions with the blood-brain and blood-tumor barriers. Finally, we discuss a selection of novel preclinical techniques that have the potential to enhance the delivery of therapeutics to brain tumors in spite of the barrier properties of the brain.
Collapse
Affiliation(s)
- Evgenii Belykh
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Kurt V. Shaffer
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Chaoqun Lin
- Department of Neurosurgery, School of Medicine, Southeast University, Nanjing, China
| | - Vadim A. Byvaltsev
- Department of Neurosurgery, Irkutsk State Medical University, Irkutsk, Russia
| | - Mark C. Preul
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Hoffman A, Atreya R, Rath T, Neurath MF. Use of Fluorescent Dyes in Endoscopy and Diagnostic Investigation. Visc Med 2020; 36:95-103. [PMID: 32355666 DOI: 10.1159/000506241] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 12/25/2022] Open
Abstract
Background The advancement of innovative endoscopic technology in terms of improving the visualization of the mucosa has been of significant benefit. Summary Advancements in image resolution, software processing, and optical filter technology have resulted in several techniques complemental to traditional white light endoscopy. These new techniques provide a real-time optical diagnosis as well as virtual histology of detected lesions. Optical molecular imaging permits a functional assessment within cells. Key Message Optical molecular imaging provides an understanding of cellular processes and permits validation of the specificity of fluorescent tracers and the possibility of quantifying the signal.
Collapse
Affiliation(s)
- Arthur Hoffman
- Department of Internal Medicine III, Clinic Aschaffenburg-Alzenau, Aschaffenburg, Germany
| | - Raja Atreya
- First Department of Medicine, Friedrich Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Timo Rath
- First Department of Medicine, Friedrich Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | - Markus F Neurath
- First Department of Medicine, Friedrich Alexander University Erlangen-Nuernberg, Erlangen, Germany
| |
Collapse
|
13
|
Near-Infrared Molecular Imaging of Glioblastoma by Miltuximab ®-IRDye800CW as a Potential Tool for Fluorescence-Guided Surgery. Cancers (Basel) 2020; 12:cancers12040984. [PMID: 32316186 PMCID: PMC7226459 DOI: 10.3390/cancers12040984] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/07/2020] [Accepted: 04/12/2020] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive tumors and its 5-year survival is approximately 5%. Fluorescence-guided surgery (FGS) improves the extent of resection and leads to better prognosis. Molecular near-infrared (NIR) imaging appears to outperform conventional FGS, however, novel molecular targets need to be identified in GBM. Proteoglycan glypican-1 (GPC-1) is believed to be such a target as it is highly expressed in GBM and is associated with poor prognosis. We hypothesize that an anti-GPC-1 antibody, Miltuximab®, conjugated with the NIR dye, IRDye800CW (IR800), can specifically accumulate in a GBM xenograft and provide high-contrast in vivo fluorescent imaging in rodents following systemic administration. Miltuximab® was conjugated with IR800 and intravenously administered to BALB/c nude mice bearing a subcutaneous U-87 GBM hind leg xenograft. Specific accumulation of Miltuximab®-IR800 in subcutaneous xenograft tumor was detected 24 h later using an in vivo fluorescence imager. The conjugate did not cause any adverse events in mice and caused strong fluorescence of the tumor with tumor-to-background ratio (TBR) reaching 10.1 ± 2.8. The average TBR over the 10-day period was 5.8 ± 0.6 in mice injected with Miltuximab®-IR800 versus 2.4 ± 0.1 for the control group injected with IgG-IR800 (p = 0.001). Ex vivo assessment of Miltuximab®-IR800 biodistribution confirmed its highly specific accumulation in the tumor. The results of this study confirm that Miltuximab®-IR800 holds promise for intraoperative fluorescence molecular imaging of GBM and warrants further studies.
Collapse
|
14
|
Oelschlägel M, Meyer T, Morgenstern U, Wahl H, Gerber J, Reiß G, Koch E, Steiner G, Kirsch M, Schackert G, Sobottka SB. Mapping of language and motor function during awake neurosurgery with intraoperative optical imaging. Neurosurg Focus 2020; 48:E3. [PMID: 32006940 DOI: 10.3171/2019.11.focus19759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/15/2019] [Indexed: 11/06/2022]
Abstract
Intraoperative optical imaging (IOI) is a marker-free, contactless, and noninvasive imaging technique that is able to visualize metabolic changes of the brain surface following neuronal activation. Although it has been used in the past mainly for the identification of functional brain areas under general anesthesia, the authors investigated the potential of the method during awake surgery. Measurements were performed in 10 patients who underwent resection of lesions within or adjacent to cortical language or motor sites. IOI was applied in 3 different scenarios: identification of motor areas by using finger-tapping tasks, identification of language areas by using speech tasks (overt and silent speech), and a novel approach-the application of IOI as a feedback tool during direct electrical stimulation (DES) mapping of language. The functional maps, which were calculated from the IOI data (activity maps), were qualitatively compared with the functional MRI (fMRI) and the electrophysiological testing results during the surgical procedure to assess their potential benefit for surgical decision-making.The results reveal that the intraoperative identification of motor sites with IOI in good agreement with the preoperatively acquired fMRI and the intraoperative electrophysiological measurements is possible. Because IOI provides spatially highly resolved maps with minimal additional hardware effort, the application of the technique for motor site identification seems to be beneficial in awake procedures. The identification of language processing sites with IOI was also possible, but in the majority of cases significant differences between fMRI, IOI, and DES were visible, and therefore according to the authors' findings the IOI results are too unspecific to be useful for intraoperative decision-making with respect to exact language localization. For this purpose, DES mapping will remain the method of choice.Nevertheless, the IOI technique can provide additional value during the language mapping procedure with DES. Using a simple difference imaging approach, the authors were able to visualize and calculate the spatial extent of activation for each stimulation. This might enable surgeons in the future to optimize the mapping process. Additionally, differences between tumor and nontumor stimulation sites were observed with respect to the spatial extent of the changes in cortical optical properties. These findings provide further evidence that the method allows the assessment of the functional state of neurovascular coupling and is therefore suited for the delineation of pathologically altered tissue.
Collapse
Affiliation(s)
- Martin Oelschlägel
- 1Clinical Sensoring and Monitoring, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden
| | - Tobias Meyer
- 2ABX-CRO Advanced Pharmaceutical Services Forschungsgesellschaft mbH, Dresden
| | - Ute Morgenstern
- 3Institute of Biomedical Engineering, Faculty of Electrical and Computer Engineering, Technische Universität Dresden
| | - Hannes Wahl
- 4Institute and Polyclinic of Diagnostic and Interventional Neuroradiology, Carl Gustav Carus University Hospital, Technische Universität Dresden
| | - Johannes Gerber
- 4Institute and Polyclinic of Diagnostic and Interventional Neuroradiology, Carl Gustav Carus University Hospital, Technische Universität Dresden
| | - Gilfe Reiß
- 6Department of Neurosurgery, Carl Gustav Carus University Hospital, Technische Universität Dresden, Saxony, Germany
| | - Edmund Koch
- 1Clinical Sensoring and Monitoring, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden
| | - Gerald Steiner
- 1Clinical Sensoring and Monitoring, Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden
| | - Matthias Kirsch
- 5Department of Neurosurgery, Asklepios Kliniken Schildautal Seesen; and
| | - Gabriele Schackert
- 6Department of Neurosurgery, Carl Gustav Carus University Hospital, Technische Universität Dresden, Saxony, Germany
| | - Stephan B Sobottka
- 6Department of Neurosurgery, Carl Gustav Carus University Hospital, Technische Universität Dresden, Saxony, Germany
| |
Collapse
|
15
|
Lee S, Park WY, Chang H, Kim B, Jang WH, Kim S, Shin Y, Kim MJ, Lee KH, Kim EH, Chung E, Kim KH. Fast and sensitive delineation of brain tumor with clinically compatible moxifloxacin labeling and confocal microscopy. JOURNAL OF BIOPHOTONICS 2020; 13:e201900197. [PMID: 31368257 DOI: 10.1002/jbio.201900197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/16/2019] [Accepted: 07/29/2019] [Indexed: 05/09/2023]
Abstract
Delineation of brain tumor margins during surgery is critical to maximize tumor removal while preserving normal brain tissue to obtain optimal clinical outcomes. Although various imaging methods have been developed, they have limitations to be used in clinical practice. We developed a high-speed cellular imaging method by using clinically compatible moxifloxacin and confocal microscopy for sensitive brain tumor detection and delineation. Moxifloxacin is a Food and Drug Administration (FDA) approved antibiotic and was used as a cell labeling agent through topical administration. Its strong fluorescence at short visible excitation wavelengths allowed video-rate cellular imaging. Moxifloxacin-based confocal microscopy (MBCM) was characterized in normal mouse brain specimens and visualized their cytoarchitecture clearly. Then, MBCM was applied to both brain tumor murine models and two malignant human brain tumors of glioblastoma and metastatic cancer. MBCM detected tumors in all the specimens by visualizing dense and irregular cell distributions, and tumor margins were easily delineated based on the cytoarchitecture. An image analysis method was developed for automated detection and delineation. MBCM demonstrated sensitive delineation of brain tumors through cytoarchitecture visualization and would have potentials for human applications, such as a surgery-guiding method for tumor removal.
Collapse
Affiliation(s)
- Seunghun Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Won Yeong Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Hoonchul Chang
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Bumju Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Won Hyuk Jang
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Seonghan Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Younghoon Shin
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Myoung Joon Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Kyung Hwa Lee
- Department of Pathology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun-gun, Jeonnam, Republic of Korea
| | - Eui Hyun Kim
- Department of Neurosurgery, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| | - Euiheon Chung
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Ki Hean Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| |
Collapse
|
16
|
Zhang DY, Singhal S, Lee JYK. Optical Principles of Fluorescence-Guided Brain Tumor Surgery: A Practical Primer for the Neurosurgeon. Neurosurgery 2019; 85:312-324. [PMID: 30085129 DOI: 10.1093/neuros/nyy315] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 06/18/2018] [Indexed: 01/21/2023] Open
Abstract
Fluorescence-guided surgery is a rapidly growing field that has produced some of the most important innovations in surgical oncology in the past decade. These intraoperative imaging technologies provide information distinguishing tumor tissue from normal tissue in real time as the surgery proceeds and without disruption of the workflow. Many of these fluorescent tracers target unique molecular or cellular features of tumors, which offers the opportunity for identifying pathology with high precision to help surgeons achieve their primary objective of a maximal safe resection. As novel fluorophores and fluorescent probes emerge from preclinical development, a practical understanding of the principles of fluorescence remains critical for evaluating the clinical utility of these agents and identifying opportunities for further innovation. In this review, we provide an "in-text glossary" of the fundamental principles of fluorescence with examples of direct applications to fluorescence-guided brain surgery. We offer a detailed discussion of the various advantages and limitations of the most commonly used intraoperative imaging agents, including 5-aminolevulinic acid, indocyanine green, and fluorescein, with a particular focus on the photophysical properties of these specific agents as they provide a framework through which to understand the new agents that are entering clinical trials. To this end, we conclude with a survey of the fluorescent properties of novel agents that are currently undergoing or will soon enter clinical trials for the intraoperative imaging of brain tumors.
Collapse
Affiliation(s)
- Daniel Y Zhang
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Cho SS, Salinas R, Lee JYK. Indocyanine-Green for Fluorescence-Guided Surgery of Brain Tumors: Evidence, Techniques, and Practical Experience. Front Surg 2019; 6:11. [PMID: 30915339 PMCID: PMC6422908 DOI: 10.3389/fsurg.2019.00011] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 02/19/2019] [Indexed: 01/10/2023] Open
Abstract
The primary treatment for brain tumors often involves surgical resection for diagnosis, relief of mass effect, and prolonged survival. In neurosurgery, it is of utmost importance to achieve maximal safe resection while minimizing iatrogenic neurologic deficit. Thus, neurosurgeons often rely on extra tools in the operating room, such as neuronavigation, intraoperative magnetic resonance imaging, and/or intraoperative rapid pathology. However, these tools can be expensive, not readily available, time-consuming, and/or inaccurate. Recently, fluorescence-guided surgery has emerged as a cost-effective method to accurately visualize neoplastic areas in real-time to guide resection. Currently, 5-aminolevulinic-acid (5-ALA) remains the only fluorophore that has been approved specifically for fluorescence-guided tumor resection. Its use has demonstrated improved resection rates and prolonged progression-free survival. However, protoporphyrin-IX, the metabolic product of 5-ALA that accumulates in neoplastic cells, fluoresces in the visible-light range, which suffers from limited tissue penetration and significant auto-fluorescence. Near-infrared fluorescence, on the other hand, overcomes these problems with ease. Since 2012, researchers at our institution have developed a novel technique using indocyanine-green, which is a well-known near-infrared fluorophore used traditionally for angiography. This Second-Window-ICG (SWIG) technique takes advantage of the increased endothelial permeability in peritumoral tissue, which allows indocyanine-green to accumulate in these areas for intraoperative visualization of the tumor. SWIG has demonstrated utility in gliomas, meningiomas, metastases, pituitary adenomas, chordomas, and craniopharyngiomas. The main benefits of SWIG stem from its highly sensitive detection of neoplastic tissue in a wide variety of intracranial pathologies in real-time, which can help neurosurgeons both during surgical resections and in stereotactic biopsies. In this review of this novel technique, we summarize the development and mechanism of action of SWIG, provide evidence for its benefits, and discuss its limitations. Finally, for those interested in near-infrared fluorescence-guided surgery, we provide suggestions for maximizing the benefits while minimizing the limitations of SWIG based on our own experience thus far.
Collapse
Affiliation(s)
- Steve S Cho
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.,Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - Ryan Salinas
- Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| | - John Y K Lee
- Department of Neurosurgery at the Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
18
|
Watson JR, Martirosyan N, Lemole GM, Trouard TP, Romanowski M. Intraoperative brain tumor resection with indocyanine green using augmented microscopy. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-4. [PMID: 30251491 PMCID: PMC6170140 DOI: 10.1117/1.jbo.23.9.090501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/07/2018] [Indexed: 06/08/2023]
Abstract
Treatment outcomes for brain cancer have seen dismal improvements over the last two decades as evident in available statistical data. Efforts to address this challenge include development of near-infrared contrast agents for improvements in diagnostic and therapeutic modalities. This creates a need for imaging technologies that can support the intraoperative use of such agents. Here, we report implementation of a recently introduced augmented microscope in combination with indocyanine green (ICG), a near-infrared contrast agent, for surgical image guidance of a glioma resection in a rat model. Luc-C6 cells were implanted in rats in the left-frontal lobe and grown for 22 days. Surgical resection was performed by a neurosurgeon using the augmented microscope with ICG contrast. ICG accumulated in the tumor tissue due to enhanced permeation and retention from the compromised blood-brain barrier. Videos and images were acquired to evaluate image quality and resection margins. The augmented microscope highlighted tumor tissue regions via visualization of ICG fluorescence and was capable of guiding the rat glioma resection.
Collapse
Affiliation(s)
- Jeffrey R. Watson
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Nikolay Martirosyan
- University of Arizona, Department of Surgery, Division of Neurosurgery, Tucson, Arizona, United States
| | - G. Michael Lemole
- University of Arizona, Department of Surgery, Division of Neurosurgery, Tucson, Arizona, United States
| | - Theodore P. Trouard
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| | - Marek Romanowski
- University of Arizona, Department of Biomedical Engineering, Tucson, Arizona, United States
| |
Collapse
|
19
|
Hu S, Kang H, Baek Y, El Fakhri G, Kuang A, Choi HS. Real-Time Imaging of Brain Tumor for Image-Guided Surgery. Adv Healthc Mater 2018; 7:e1800066. [PMID: 29719137 PMCID: PMC6105507 DOI: 10.1002/adhm.201800066] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/22/2018] [Indexed: 02/05/2023]
Abstract
The completion of surgical resection is a key prognostic factor in brain tumor treatment. This requires surgeons to identify residual tumors in theater as well as to margin the proximity of the tumor to adjacent normal tissue. Subjective assessments, such as texture palpation or visual tissue differences, are commonly used by oncology surgeons during resection to differentiate cancer lesions from normal tissue, which can potentially result in either an incomplete tumor resection, or accidental removal of normal tissue. Moreover, malignant brain tumors are even more difficult to distinguish from normal brain tissue, and resecting noncancerous tissue may create neurological defects after surgery. To optimize the resection margin in brain tumors, a variety of intraoperative guidance techniques are developed, such as neuronavigation, magnetic resonance imaging, ultrasound, Raman spectroscopy, and optical fluorescence imaging. When combined with appropriate contrast agents, optical fluorescence imaging can provide the neurosurgeon real-time image guidance to improve resection completeness and to decrease surgical complications.
Collapse
Affiliation(s)
- Shuang Hu
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yoonji Baek
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Anren Kuang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
20
|
Lee JYK, Pierce JT, Thawani JP, Zeh R, Nie S, Martinez-Lage M, Singhal S. Near-infrared fluorescent image-guided surgery for intracranial meningioma. J Neurosurg 2018; 128:380-390. [PMID: 28387632 PMCID: PMC10985532 DOI: 10.3171/2016.10.jns161636] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Meningiomas are the most common primary tumor of the central nervous system. Complete resection can be curative, but intraoperative identification of dural tails and tumor remnants poses a clinical challenge. Given data from preclinical studies and previous clinical trials, the authors propose a novel method of localizing tumor tissue and identifying residual disease at the margins via preoperative systemic injection of a near-infrared (NIR) fluorescent contrast dye. This technique, what the authors call "second-window indocyanine green" (ICG), relies on the visualization of ICG approximately 24 hours after intravenous injection. METHODS Eighteen patients were prospectively identified and received 5 mg/kg of second-window ICG the day prior to surgery. An NIR camera was used to localize the tumor prior to resection and to inspect the margins following standard resection. The signal to background ratio (SBR) of the tumor to the normal brain parenchyma was measured in triplicate. Gross tumor and margin specimens were qualitatively reported with respect to fluorescence. Neuropathological diagnosis served as the reference gold standard to calculate the sensitivity and specificity of the imaging technique. RESULTS Eighteen patients harbored 15 WHO Grade I and 3 WHO Grade II meningiomas. Near-infrared visualization during surgery ranged from 18 to 28 hours (mean 23 hours) following second-window ICG infusion. Fourteen of the 18 tumors demonstrated a markedly elevated SBR of 5.6 ± 1.7 as compared with adjacent brain parenchyma. Four of the 18 patients showed an inverse pattern of NIR signal, that is, stronger in the adjacent normal brain than in the tumor (SBR 0.31 ± 0.1). The best predictor of inversion was time from injection, as the patients who were imaged earlier were more likely to demonstrate an appropriate SBR. The second-window ICG technique demonstrated a sensitivity of 96.4%, specificity of 38.9%, positive predictive value of 71.1%, and a negative predictive value of 87.5% for tumor. CONCLUSIONS Systemic injection of NIR second-window ICG the day before surgery can be used to visualize meningiomas intraoperatively. Intraoperative NIR imaging provides higher sensitivity in identifying meningiomas than the unassisted eye. In this study, 14 of the 18 patients with meningioma demonstrated a strong SBR compared with adjacent brain. In the future, reducing the time interval from dye injection to intraoperative imaging may improve fluorescence at the margins, though this approach requires further investigation. Clinical trial registration no.: NCT02280954 ( clincialtrials.gov ).
Collapse
Affiliation(s)
- John Y. K. Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania
- Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - John T. Pierce
- Department of Neurosurgery, Hospital of the University of Pennsylvania
| | - Jayesh P. Thawani
- Department of Neurosurgery, Hospital of the University of Pennsylvania
| | - Ryan Zeh
- Department of Neurosurgery, Hospital of the University of Pennsylvania
| | - Shuming Nie
- Department of Chemistry, Emory University School of Medicine, Atlanta, Georgia
| | | | - Sunil Singhal
- Department of Surgery, Hospital of the University of Pennsylvania
- Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Lee JYK, Thawani JP, Pierce J, Zeh R, Martinez-Lage M, Chanin M, Venegas O, Nims S, Learned K, Keating J, Singhal S. Intraoperative Near-Infrared Optical Imaging Can Localize Gadolinium-Enhancing Gliomas During Surgery. Neurosurgery 2017; 79:856-871. [PMID: 27741220 DOI: 10.1227/neu.0000000000001450] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Although real-time localization of gliomas has improved with intraoperative image guidance systems, these tools are limited by brain shift, surgical cavity deformation, and expense. OBJECTIVE To propose a novel method to perform near-infrared (NIR) imaging during glioma resections based on preclinical and clinical investigations, in order to localize tumors and to potentially identify residual disease. METHODS Fifteen patients were identified and administered a Food and Drug Administration-approved, NIR contrast agent (Second Window indocyanine green [ICG], 5 mg/kg) before surgical resection. An NIR camera was utilized to localize the tumor before resection and to visualize surgical margins following resection. Neuropathology and magnetic resonance imaging data were used to assess the accuracy and precision of NIR fluorescence in identifying tumor tissue. RESULTS NIR visualization of 15 gliomas (10 glioblastoma multiforme, 1 anaplastic astrocytoma, 2 low-grade astrocytoma, 1 juvenile pilocytic astrocytoma, and 1 ganglioglioma) was performed 22.7 hours (mean) after intravenous injection of ICG. During surgery, 12 of 15 tumors were visualized with the NIR camera. The mean signal-to-background ratio was 9.5 ± 0.8 and fluorescence was noted through the dura to a maximum parenchymal depth of 13 mm. The best predictor of positive fluorescence was enhancement on T1-weighted imaging; this correlated with signal-to-background ratio (P = .03). Nonenhancing tumors did not demonstrate NIR fluorescence. Using pathology as the gold standard, the technique demonstrated a sensitivity of 98% and specificity of 45% to identify tumor in gadolinium-enhancing specimens (n = 71). CONCLUSION With the use of Second Window ICG, gadolinium-enhancing tumors can be localized through brain parenchyma intraoperatively. Its utility for margin detection is promising but limited by lower specificity. ABBREVIATIONS 5-ALA, 5-aminolevulinic acidEPR, enhanced permeability and retentionFDA, Food and Drug AdministrationGBM, glioblastomaICG, indocyanine greenNIR, near-infraredNPV, negative predictive valuePPV, positive predictive valueROC, receiver operating characteristicROI, region of interestSBR, signal-to-background ratioWHO, World Health Organization.
Collapse
Affiliation(s)
- John Y K Lee
- *Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania; ‡Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania; §Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania; ¶Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Sun C, Du W, Wang B, Yao Z, Liu J, Wang J, Xie W, Wu T, Fan Y, Yang H. Synthesis of a new deoxyglucose derivative modified near-infrared fluorescent probe for tumor diagnosis. Biochem Biophys Res Commun 2017; 488:340-347. [PMID: 28499871 DOI: 10.1016/j.bbrc.2017.05.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 10/19/2022]
Abstract
Malignant neoplasms exhibit an elevated rate of glycolysis and a high demand for glucose over normal cells. This characteristic can be exploited for in vivo imaging and tumor targeting examined. In this manuscript, we describe the synthesis of near-infrared (NIR) fluorochrome IR-822-labeled 2-amino-2-deoxy-d-glucose (DG) for optical imaging of tumors in mice. NIR fluorescent dye IR-820 was subsequently conjugated with 3-Mercaptopropionic acid and 2-amino-2-deoxy-d-glucose to form IR-822-DG. The cell experiments and acute toxicity studies demonstrated the low toxicity of IR-822-DG to normal cells/tissues. The dynamic behavior and targeting ability of IR-822-DG in normal mice was investigated with a NIR fluorescence imaging system. The in vitro and in vivo tumor targeting capabilities of IR-822-DG were evaluated in tumor cells and tumor bearing mice, respectively. Results demonstrated that IR-822-DG actively and efficiently accumulated at the site of the tumor. The probe also exhibited good photostability and excellent cell membrane permeability. The study indicates the broad applicability of IR-822-DG for tumors diagnosis, especially in the glucose-related pathologies.
Collapse
Affiliation(s)
- Chunlong Sun
- Shandong Provincial Key Laboratory of Eco-environmental Science for Yellow River Delta, Binzhou University, Binzhou 256600, China; School of Bioengineering, Binzhou University, Binzhou 256600, China.
| | - Wen Du
- Shandong Provincial Key Laboratory of Eco-environmental Science for Yellow River Delta, Binzhou University, Binzhou 256600, China; School of Bioengineering, Binzhou University, Binzhou 256600, China.
| | - Baoqin Wang
- School of Bioengineering, Binzhou University, Binzhou 256600, China
| | - Zhigang Yao
- School of Bioengineering, Binzhou University, Binzhou 256600, China
| | - Junhua Liu
- Shandong Provincial Key Laboratory of Eco-environmental Science for Yellow River Delta, Binzhou University, Binzhou 256600, China
| | - Jun Wang
- School of Bioengineering, Binzhou University, Binzhou 256600, China
| | - Wenjun Xie
- School of Bioengineering, Binzhou University, Binzhou 256600, China
| | - Tao Wu
- Shandong Provincial Engineering and Technology Research Center for Wild Plant Resources Development and Application of Yellow River Delta, School of Bioengineering, Binzhou University, Binzhou 256600, China
| | - Yanhui Fan
- School of Bioengineering, Binzhou University, Binzhou 256600, China
| | - Hongjun Yang
- Shandong Provincial Key Laboratory of Eco-environmental Science for Yellow River Delta, Binzhou University, Binzhou 256600, China
| |
Collapse
|
23
|
|
24
|
Senders JT, Muskens IS, Schnoor R, Karhade AV, Cote DJ, Smith TR, Broekman MLD. Agents for fluorescence-guided glioma surgery: a systematic review of preclinical and clinical results. Acta Neurochir (Wien) 2017; 159:151-167. [PMID: 27878374 PMCID: PMC5177668 DOI: 10.1007/s00701-016-3028-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/09/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Fluorescence-guided surgery (FGS) is a technique used to enhance visualization of tumor margins in order to increase the extent of tumor resection in glioma surgery. In this paper, we systematically review all clinically tested fluorescent agents for application in FGS for glioma and all preclinically tested agents with the potential for FGS for glioma. METHODS We searched the PubMed and Embase databases for all potentially relevant studies through March 2016. We assessed fluorescent agents by the following outcomes: rate of gross total resection (GTR), overall and progression-free survival, sensitivity and specificity in discriminating tumor and healthy brain tissue, tumor-to-normal ratio of fluorescent signal, and incidence of adverse events. RESULTS The search strategy resulted in 2155 articles that were screened by titles and abstracts. After full-text screening, 105 articles fulfilled the inclusion criteria evaluating the following fluorescent agents: 5-aminolevulinic acid (5-ALA) (44 studies, including three randomized control trials), fluorescein (11), indocyanine green (five), hypericin (two), 5-aminofluorescein-human serum albumin (one), endogenous fluorophores (nine) and fluorescent agents in a pre-clinical testing phase (30). Three meta-analyses were also identified. CONCLUSIONS 5-ALA is the only fluorescent agent that has been tested in a randomized controlled trial and results in an improvement of GTR and progression-free survival in high-grade gliomas. Observational cohort studies and case series suggest similar outcomes for FGS using fluorescein. Molecular targeting agents (e.g., fluorophore/nanoparticle labeled with anti-EGFR antibodies) are still in the pre-clinical phase, but offer promising results and may be valuable future alternatives.
Collapse
Affiliation(s)
- Joeky T Senders
- Department of Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ivo S Muskens
- Department of Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Rosalie Schnoor
- Department of Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Aditya V Karhade
- Department of Neurosurgery, Cushing Neurosurgery Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, 15 Francis Street, Boston, MA, 02115, USA
| | - David J Cote
- Department of Neurosurgery, Cushing Neurosurgery Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, 15 Francis Street, Boston, MA, 02115, USA
| | - Timothy R Smith
- Department of Neurosurgery, Cushing Neurosurgery Outcomes Center, Brigham and Women's Hospital, Harvard Medical School, 15 Francis Street, Boston, MA, 02115, USA
| | - Marike L D Broekman
- Department of Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
25
|
Valdés PA, Roberts DW, Lu FK, Golby A. Optical technologies for intraoperative neurosurgical guidance. Neurosurg Focus 2016; 40:E8. [PMID: 26926066 DOI: 10.3171/2015.12.focus15550] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Biomedical optics is a broadly interdisciplinary field at the interface of optical engineering, biophysics, computer science, medicine, biology, and chemistry, helping us understand light-tissue interactions to create applications with diagnostic and therapeutic value in medicine. Implementation of biomedical optics tools and principles has had a notable scientific and clinical resurgence in recent years in the neurosurgical community. This is in great part due to work in fluorescence-guided surgery of brain tumors leading to reports of significant improvement in maximizing the rates of gross-total resection. Multiple additional optical technologies have been implemented clinically, including diffuse reflectance spectroscopy and imaging, optical coherence tomography, Raman spectroscopy and imaging, and advanced quantitative methods, including quantitative fluorescence and lifetime imaging. Here we present a clinically relevant and technologically informed overview and discussion of some of the major clinical implementations of optical technologies as intraoperative guidance tools in neurosurgery.
Collapse
Affiliation(s)
- Pablo A Valdés
- Departments of 1 Neurosurgery and.,Department of Neurosurgery, Harvard Medical School, Boston Children's Hospital, Boston
| | - David W Roberts
- Section of Neurosurgery, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | | - Alexandra Golby
- Departments of 1 Neurosurgery and.,Radiology, and.,Dana Farber Cancer Institute, Harvard Medical School, Brigham and Women's Hospital
| |
Collapse
|
26
|
Vieira DB, Gamarra LF. Getting into the brain: liposome-based strategies for effective drug delivery across the blood-brain barrier. Int J Nanomedicine 2016; 11:5381-5414. [PMID: 27799765 PMCID: PMC5077137 DOI: 10.2147/ijn.s117210] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review summarizes articles that have been reported in literature on liposome-based strategies for effective drug delivery across the blood–brain barrier. Due to their unique physicochemical characteristics, liposomes have been widely investigated for their application in drug delivery and in vivo bioimaging for the treatment and/or diagnosis of neurological diseases, such as Alzheimer’s, Parkinson’s, stroke, and glioma. Several strategies have been used to deliver drug and/or imaging agents to the brain. Covalent ligation of such macromolecules as peptides, antibodies, and RNA aptamers is an effective method for receptor-targeting liposomes, which allows their blood–brain barrier penetration and/or the delivery of their therapeutic molecule specifically to the disease site. Additionally, methods have been employed for the development of liposomes that can respond to external stimuli. It can be concluded that the development of liposomes for brain delivery is still in its infancy, although these systems have the potential to revolutionize the ways in which medicine is administered.
Collapse
Affiliation(s)
| | - Lionel F Gamarra
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| |
Collapse
|
27
|
Belykh E, Martirosyan NL, Yagmurlu K, Miller EJ, Eschbacher JM, Izadyyazdanabadi M, Bardonova LA, Byvaltsev VA, Nakaji P, Preul MC. Intraoperative Fluorescence Imaging for Personalized Brain Tumor Resection: Current State and Future Directions. Front Surg 2016; 3:55. [PMID: 27800481 PMCID: PMC5066076 DOI: 10.3389/fsurg.2016.00055] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/12/2016] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Fluorescence-guided surgery is one of the rapidly emerging methods of surgical "theranostics." In this review, we summarize current fluorescence techniques used in neurosurgical practice for brain tumor patients as well as future applications of recent laboratory and translational studies. METHODS Review of the literature. RESULTS A wide spectrum of fluorophores that have been tested for brain surgery is reviewed. Beginning with a fluorescein sodium application in 1948 by Moore, fluorescence-guided brain tumor surgery is either routinely applied in some centers or is under active study in clinical trials. Besides the trinity of commonly used drugs (fluorescein sodium, 5-aminolevulinic acid, and indocyanine green), less studied fluorescent stains, such as tetracyclines, cancer-selective alkylphosphocholine analogs, cresyl violet, acridine orange, and acriflavine, can be used for rapid tumor detection and pathological tissue examination. Other emerging agents, such as activity-based probes and targeted molecular probes that can provide biomolecular specificity for surgical visualization and treatment, are reviewed. Furthermore, we review available engineering and optical solutions for fluorescent surgical visualization. Instruments for fluorescent-guided surgery are divided into wide-field imaging systems and hand-held probes. Recent advancements in quantitative fluorescence-guided surgery are discussed. CONCLUSION We are standing on the threshold of the era of marker-assisted tumor management. Innovations in the fields of surgical optics, computer image analysis, and molecular bioengineering are advancing fluorescence-guided tumor resection paradigms, leading to cell-level approaches to visualization and resection of brain tumors.
Collapse
Affiliation(s)
- Evgenii Belykh
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Laboratory of Neurosurgery, Irkutsk Scientific Center of Surgery and Traumatology, Irkutsk, Russia
- Irkutsk State Medical University, Irkutsk, Russia
| | - Nikolay L. Martirosyan
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Kaan Yagmurlu
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Eric J. Miller
- University of Arizona College of Medicine – Phoenix, Phoenix, AZ, USA
| | - Jennifer M. Eschbacher
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Mohammadhassan Izadyyazdanabadi
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Liudmila A. Bardonova
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
- Laboratory of Neurosurgery, Irkutsk Scientific Center of Surgery and Traumatology, Irkutsk, Russia
- Irkutsk State Medical University, Irkutsk, Russia
| | - Vadim A. Byvaltsev
- Laboratory of Neurosurgery, Irkutsk Scientific Center of Surgery and Traumatology, Irkutsk, Russia
- Irkutsk State Medical University, Irkutsk, Russia
| | - Peter Nakaji
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Mark C. Preul
- Department of Neurosurgery, St. Joseph’s Hospital and Medical Center, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
28
|
De Grand AM, Frangioni JV. An Operational Near-Infrared Fluorescence Imaging System Prototype for Large Animal Surgery. Technol Cancer Res Treat 2016; 2:553-62. [PMID: 14640766 DOI: 10.1177/153303460300200607] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Near-infrared (NIR) fluorescence imaging has the potential to revolutionize human cancer surgery by providing sensitive, specific, and real-time intraoperative visualization of normal and disease processes. We have previously introduced the concept of a low-cost, safe, and easy-to-use NIR fluorescence imaging system that permits the surgeon to “see” surgical anatomy and NIR fluorescence simultaneously, non-invasively, with high spatial resolution, in real-time, and without moving parts [Nakayama et al. Molecular Imaging 1, 365–377 (2002)]. In this study, we present an operational prototype designed specifically for use during large animal surgery. Such a system serves as a foundation for future clinical studies. We discuss technical considerations, and provide details of the implementation of subsystems related to excitation light, light collection, computer, and software. Using the prototype, and the clinically available NIR fluorophore indocyanine green, we demonstrate vascular imaging in 35 kg pigs. Cancer-specific applications of this imaging system include image-guided cancer resection with real-time assessment of surgical margins, image-guided sentinel lymph node mapping, intraoperative mapping of tumor and normal vasculature, image-guided avoidance of critical structures such as nerves, and intraoperative detection of occult metastases in the surgical field. Taken together, this study describes an optical imaging system engineered for eventual translation to the clinic.
Collapse
Affiliation(s)
- A M De Grand
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, SL-B05, 330 Brookline Avenue, Boston, MA 02215, USA
| | | |
Collapse
|
29
|
Craig SEL, Wright J, Sloan AE, Brady-Kalnay SM. Fluorescent-Guided Surgical Resection of Glioma with Targeted Molecular Imaging Agents: A Literature Review. World Neurosurg 2016; 90:154-163. [PMID: 26915698 PMCID: PMC4915969 DOI: 10.1016/j.wneu.2016.02.060] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/11/2022]
Abstract
The median life expectancy after a diagnosis of glioblastoma is 15 months. Although chemotherapeutics may someday cure glioblastoma by killing the highly dispersive malignant cells, the most important contribution that clinicians can currently offer to improve survival is by maximizing the extent of resection and providing concurrent chemo-radiation, which has become standard. Strides have been made in this area with the advent and implementation of methods of improved intraoperative tumor visualization. One of these techniques, optical fluorescent imaging with targeted molecular imaging agents, allows the surgeon to view fluorescently labeled tumor tissue during surgery with the use of special microscopy, thereby highlighting where to resect and indicating when tumor-free margins have been obtained. This advantage is especially important at the difficult-to-observe margins where tumor cells infiltrate normal tissue. Targeted fluorescent agents also may be valuable for identifying tumor versus nontumor tissue. In this review, we briefly summarize nontargeted fluorescent tumor imaging agents before discussing several novel targeted fluorescent agents being developed for glioma imaging in the context of fluorescent-guided surgery or live molecular navigation. Many of these agents are currently undergoing preclinical testing. As the agents become available, however, it is necessary to understand the strengths and weaknesses of each.
Collapse
Affiliation(s)
- Sonya E L Craig
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - James Wright
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Andrew E Sloan
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA; Brain Tumor and Neuro-Oncology Center, University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Susann M Brady-Kalnay
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
30
|
Gao C, Deng ZJ, Peng D, Jin YS, Ma Y, Li YY, Zhu YK, Xi JZ, Tian J, Dai ZF, Li CH, Liang XL. Near-infrared dye-loaded magnetic nanoparticles as photoacoustic contrast agent for enhanced tumor imaging. Cancer Biol Med 2016; 13:349-359. [PMID: 27807502 PMCID: PMC5069831 DOI: 10.20892/j.issn.2095-3941.2016.0048] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Objective: Photoacoustic (PA) tomography (PAT) has attracted extensive interest because of its optical absorption contrast and ultrasonic detection. This study aims to develop a biocompatible and biodegradable PA contrast agent particularly promising for clinical applications in human body.
Methods: In this study, we presented a PA contrast agent: 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-
N-[methoxy (polyethylene glycol)] (DSPE-PEG)-coated superparamagnetic iron oxide (SPIO) nanoparticles (NPs) loaded with indocyanine green (ICG). We used ICG and SPIO NPs because both drugs are approved by the U.S. Food and Drug Administration. Given the strong absorption of near-infrared laser pulses, SPIO@DSPE-PEG/ICG NPs with a uniform diameter of ~28 nm could significantly enhance PA signals.
Results: We demonstrated the contrast enhancement of these NPs in phantom and animal experiments, in which the
in vivo circulation time of SPIO@DSPE-PEG/ICG NPs was considerably longer than that of free ICG. These novel NPs also displayed a high efficiency of tumor targeting.
Conclusions: SPIO@DSPE-PEG/ICG NPs are promising PAT contrast agents for clinical applications.
Collapse
Affiliation(s)
- Chuang Gao
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Zi-Jian Deng
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Dong Peng
- Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yu-Shen Jin
- Nanomedicine and Biosensor Laboratory, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yan Ma
- Nanomedicine and Biosensor Laboratory, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yan-Yan Li
- Nanomedicine and Biosensor Laboratory, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yu-Kun Zhu
- Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jian-Zhong Xi
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Jie Tian
- Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhi-Fei Dai
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Chang-Hui Li
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Xiao-Long Liang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
31
|
Guyotat J, Pallud J, Armoiry X, Pavlov V, Metellus P. 5-Aminolevulinic Acid-Protoporphyrin IX Fluorescence-Guided Surgery of High-Grade Gliomas: A Systematic Review. Adv Tech Stand Neurosurg 2016:61-90. [PMID: 26508406 DOI: 10.1007/978-3-319-21359-0_3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The current first-line treatment of malignant gliomas consists in surgical resection (if possible) as large as possible. The existing tools don't permit to identify the limits of tumor infiltration, which goes beyond the zone of contrast enhancement on MRI. The fluorescence-guided malignant gliomas surgery was started 15 years ago and had become a standard of care in many countries. The technique is based on fluorescent molecule revelation using the filters, positioned within the surgical microscope. The fluorophore, protoporphyrin IX (PpIX), is converted in tumoral cells from 5-aminolevulinic acid (5-ALA), given orally before surgery. Many studies have shown that the ratio of gross total resections was higher if the fluorescence technique was used. The fluorescence signal intensity is correlated to the cell density and the PpIX concentration. The current method has a very high specificity but still lower sensibility, particularly regarding the zones with poor tumoral infiltration. This book reviews the principles of the technique and the results (extent of resection and survival).
Collapse
Affiliation(s)
- Jacques Guyotat
- Department of Neurosurgery, Neurological Hospital, Lyon, France.
| | - Johan Pallud
- Department of Neurosurgery, Sainte Anne Hospital, Paris, France.
- Paris Descartes University, Paris, France.
| | - Xavier Armoiry
- Délégation à la recherche clinique et à l'innovation, cellule innovation Hospices Civils de, Lyon, France.
| | - Vladislav Pavlov
- Department of Neurosurgery, Neurological Hospital, Lyon, France.
| | - Philippe Metellus
- Department of Neurosurgery, Timone Hospital, Marseille, France.
- University Aix Marseille, Marseille, France.
| |
Collapse
|
32
|
Holt D, Parthasarathy AB, Okusanya O, Keating J, Venegas O, Deshpande C, Karakousis G, Madajewski B, Durham A, Nie S, Yodh AG, Singhal S. Intraoperative near-infrared fluorescence imaging and spectroscopy identifies residual tumor cells in wounds. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:76002. [PMID: 26160347 PMCID: PMC4497968 DOI: 10.1117/1.jbo.20.7.076002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/08/2015] [Indexed: 05/09/2023]
Abstract
Surgery is the most effective method to cure patients with solid tumors, and 50% of all cancer patients undergo resection. Local recurrences are due to tumor cells remaining in the wound, thus we explore near-infrared (NIR) fluorescence spectroscopy and imaging to identify residual cancer cells after surgery. Fifteen canines and two human patients with spontaneously occurring sarcomas underwent intraoperative imaging. During the operation, the wounds were interrogated with NIR fluorescence imaging and spectroscopy. NIR monitoring identified the presence or absence of residual tumor cells after surgery in 14/15 canines with a mean fluorescence signal-to-background ratio (SBR) of ∼16 . Ten animals showed no residual tumor cells in the wound bed (mean SBR<2 , P<0.001 ). None had a local recurrence at >1-year follow-up. In five animals, the mean SBR of the wound was >15 , and histopathology confirmed tumor cells in the postsurgical wound in four/five canines. In the human pilot study, neither patient had residual tumor cells in the wound bed, and both remain disease free at >1.5-year follow up. Intraoperative NIR fluorescence imaging and spectroscopy identifies residual tumor cells in surgical wounds. These observations suggest that NIR imaging techniques may improve tumor resection during cancer operations.
Collapse
Affiliation(s)
- David Holt
- University of Pennsylvania School of Veterinary Medicine, Department of Clinical Studies, Philadelphia, Pennsylvania 19104, United States
- Address all correspondence to: David Holt, E-mail:
| | - Ashwin B. Parthasarathy
- University of Pennsylvania, Department of Physics and Astronomy, Philadelphia, Pennsylvania 19104, United States
| | - Olugbenga Okusanya
- University of Pennsylvania School of Medicine, Department of Surgery, Philadelphia, Pennsylvania 19104, United States
| | - Jane Keating
- University of Pennsylvania School of Medicine, Department of Surgery, Philadelphia, Pennsylvania 19104, United States
| | - Ollin Venegas
- University of Pennsylvania School of Medicine, Department of Surgery, Philadelphia, Pennsylvania 19104, United States
| | - Charuhas Deshpande
- University of Pennsylvania School of Medicine, Department of Pathology, Philadelphia, Pennsylvania 19104, United States
| | - Giorgos Karakousis
- University of Pennsylvania School of Medicine, Department of Pathology, Philadelphia, Pennsylvania 19104, United States
| | - Brian Madajewski
- University of Pennsylvania School of Medicine, Department of Surgery, Philadelphia, Pennsylvania 19104, United States
| | - Amy Durham
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, Pennsylvania 19104, United States
| | - Shuming Nie
- Emory University, Departments of Biomedical Engineering and Chemistry, Atlanta, Georgia 30322, United States
| | - Arjun G. Yodh
- University of Pennsylvania, Department of Physics and Astronomy, Philadelphia, Pennsylvania 19104, United States
| | - Sunil Singhal
- University of Pennsylvania School of Medicine, Department of Surgery, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
33
|
Xi L, Jiang H. Image-guided surgery using multimodality strategy and molecular probes. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:46-60. [PMID: 26053199 DOI: 10.1002/wnan.1352] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/23/2015] [Accepted: 04/19/2015] [Indexed: 12/15/2022]
Abstract
The ultimate goal of cancer surgery is to maximize the excision of tumorous tissue with minimal damage to the collateral normal tissues, reduce the postoperative recurrence, and improve the survival rate of patients. In order to locate tumor lesions, highlight tumor margins, visualize residual disease in the surgical wound, and map potential lymph node metastasis, various imaging techniques and molecular probes have been investigated to assist surgeons to perform more complete tumor resection. Combining imaging techniques with molecular probes is particularly promising as a new approach for image-guided surgery. Considering inherent limitations of different imaging techniques and insufficient sensitivity of nonspecific molecular probes, image-guided surgery with multimodality strategy and specific molecular probes appears to be an optimal choice. In this article, we briefly describe typical imaging techniques and molecular probes followed by a focused review on the current progress of multimodal image-guided surgery with specific molecular navigation. We also discuss optimal strategy that covers all stages of image-guided surgery including preoperative scanning of tumors, intraoperative inspection of surgical bed and postoperative care of patients.
Collapse
Affiliation(s)
- Lei Xi
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu, China
| | - Hubei Jiang
- School of Physical Electronics, University of Electronic Science and Technology of China, Chengdu, China.,Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| |
Collapse
|
34
|
Parthasarathy AB, Chong SH, Moscatelli FA, Singhal S, Yodh AG. Intraoperative imaging of tumors with indo-cyanine green fluorescence with an endoscope. ACTA ACUST UNITED AC 2015. [DOI: 10.1117/12.2080298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
35
|
Missios S, Abbassy M, Vogelbaum MA, Recinos PF. Use of Image Fluorescence in the Resection of Gliomas. CURRENT SURGERY REPORTS 2014. [DOI: 10.1007/s40137-014-0076-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
36
|
Mooney MA, Zehri AH, Georges JF, Nakaji P. Laser scanning confocal endomicroscopy in the neurosurgical operating room: a review and discussion of future applications. Neurosurg Focus 2014; 36:E9. [PMID: 24484262 DOI: 10.3171/2013.11.focus13484] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Laser scanning confocal endomicroscopy (LSCE) is an emerging technology for examining brain neoplasms in vivo. While great advances have been made in macroscopic fluorescence in recent years, the ability to perform confocal microscopy in vivo expands the potential of fluorescent tumor labeling, can improve intraoperative tissue diagnosis, and provides real-time guidance for tumor resection intraoperatively. In this review, the authors highlight the technical aspects of confocal endomicroscopy and fluorophores relevant to the neurosurgeon, provide a comprehensive summary of LSCE in animal and human neurosurgical studies to date, and discuss the future directions and potential for LSCE in neurosurgery.
Collapse
|
37
|
Bu L, Shen B, Cheng Z. Fluorescent imaging of cancerous tissues for targeted surgery. Adv Drug Deliv Rev 2014; 76:21-38. [PMID: 25064553 DOI: 10.1016/j.addr.2014.07.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 05/29/2014] [Accepted: 07/10/2014] [Indexed: 12/18/2022]
Abstract
To maximize tumor excision and minimize collateral damage are the primary goals of cancer surgery. Emerging molecular imaging techniques have made "image-guided surgery" developed into "molecular imaging-guided surgery", which is termed as "targeted surgery" in this review. Consequently, the precision of surgery can be advanced from tissue-scale to molecule-scale, enabling "targeted surgery" to be a component of "targeted therapy". Evidence from numerous experimental and clinical studies has demonstrated significant benefits of fluorescent imaging in targeted surgery with preoperative molecular diagnostic screening. Fluorescent imaging can help to improve intraoperative staging and enable more radical cytoreduction, detect obscure tumor lesions in special organs, highlight tumor margins, better map lymph node metastases, and identify important normal structures intraoperatively. Though limited tissue penetration of fluorescent imaging and tumor heterogeneity are two major hurdles for current targeted surgery, multimodality imaging and multiplex imaging may provide potential solutions to overcome these issues, respectively. Moreover, though many fluorescent imaging techniques and probes have been investigated, targeted surgery remains at a proof-of-principle stage. The impact of fluorescent imaging on cancer surgery will likely be realized through persistent interdisciplinary amalgamation of research in diverse fields.
Collapse
|
38
|
Wang X, Huang SS, Heston WDW, Guo H, Wang BC, Basilion JP. Development of targeted near-infrared imaging agents for prostate cancer. Mol Cancer Ther 2014; 13:2595-606. [PMID: 25239933 DOI: 10.1158/1535-7163.mct-14-0422] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate cancer is the most common noncutaneous malignancy affecting men in North America. Radical prostatectomy remains a definitive treatment for prostate cancer. However, prostate surgeries are still performed "blindly" with the extent of tumor infiltration past the margins of the surgery only being determined postoperatively. An imaging modality that can be used during surgery is needed to help define the tumor margins. With its abundant expression in prostate cancer, prostate-specific membrane antigen (PSMA) is an ideal target for detection of prostate cancer. The purpose of this study was to develop PSMA-targeted near-infrared (NIR) optical imaging probes for intraoperative visualization of prostate cancer. We synthesized a high-affinity PSMA ligand (PSMA-1) with low molecular weight and further labeled it with commercially available NIR dyes IRDy800 and Cy5.5. PSMA-1 and PSMA-1-NIR conjugates had binding affinities better than the parent ligand Cys-CO-Glu. Selective binding was measured for each of the probes in both in vitro and in vivo studies using competitive binding and uptake studies. Interestingly, the results indicated that the pharmacokinetics of the probes was dependent of the fluorophore conjugated to the PSMA-1 ligand and varied widely. These data suggest that PSMA-targeted probes have the potential to be further developed as contrast agents for clinical intraoperative fluorescence-guided surgery.
Collapse
Affiliation(s)
- Xinning Wang
- Department of Radiology and NFCR Center for Molecular Imaging, Case Western Reserve University, Cleveland, Ohio
| | - Steve S Huang
- Department of Nuclear Medicine, Cleveland Clinic, Cleveland, Ohio
| | | | - Hong Guo
- Departments of Medicine, Pharmacology and Oncology, MetroHealth Campus, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Bing-Cheng Wang
- Departments of Medicine, Pharmacology and Oncology, MetroHealth Campus, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - James P Basilion
- Department of Radiology and NFCR Center for Molecular Imaging, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
39
|
Zehri AH, Ramey W, Georges JF, Mooney MA, Martirosyan NL, Preul MC, Nakaji P. Neurosurgical confocal endomicroscopy: A review of contrast agents, confocal systems, and future imaging modalities. Surg Neurol Int 2014; 5:60. [PMID: 24872922 PMCID: PMC4033764 DOI: 10.4103/2152-7806.131638] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 02/13/2014] [Indexed: 01/15/2023] Open
Abstract
Background: The clinical application of fluorescent contrast agents (fluorescein, indocyanine green, and aminolevulinic acid) with intraoperative microscopy has led to advances in intraoperative brain tumor imaging. Their properties, mechanism of action, history of use, and safety are analyzed in this report along with a review of current laser scanning confocal endomicroscopy systems. Additional imaging modalities with potential neurosurgical utility are also analyzed. Methods: A comprehensive literature search was performed utilizing PubMed and key words: In vivo confocal microscopy, confocal endomicroscopy, fluorescence imaging, in vivo diagnostics/neoplasm, in vivo molecular imaging, and optical imaging. Articles were reviewed that discussed clinically available fluorophores in neurosurgery, confocal endomicroscopy instrumentation, confocal microscopy systems, and intraoperative cancer diagnostics. Results: Current clinically available fluorescent contrast agents have specific properties that provide microscopic delineation of tumors when imaged with laser scanning confocal endomicroscopes. Other imaging modalities such as coherent anti-Stokes Raman scattering (CARS) microscopy, confocal reflectance microscopy, fluorescent lifetime imaging (FLIM), two-photon microscopy, and second harmonic generation may also have potential in neurosurgical applications. Conclusion: In addition to guiding tumor resection, intraoperative fluorescence and microscopy have the potential to facilitate tumor identification and complement frozen section analysis during surgery by providing real-time histological assessment. Further research, including clinical trials, is necessary to test the efficacy of fluorescent contrast agents and optical imaging instrumentation in order to establish their role in neurosurgery.
Collapse
Affiliation(s)
- Aqib H Zehri
- Neurosurgery Research Laboratory, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Wyatt Ramey
- Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Joseph F Georges
- Neurosurgery Research Laboratory, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA ; School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Michael A Mooney
- Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Nikolay L Martirosyan
- Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA ; Division of Neurosurgery, Department of Surgery, The University of Arizona, Tucson, AZ, Arizona, USA
| | - Mark C Preul
- Neurosurgery Research Laboratory, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Peter Nakaji
- Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
40
|
De la Garza-Ramos R, Bydon M, Macki M, Huang J, Tamargo RJ, Bydon A. Fluorescent techniques in spine surgery. Neurol Res 2014; 36:928-38. [DOI: 10.1179/1743132814y.0000000340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Synthesis of a near-infrared fluorescent probe and its application in imaging of MCF-7 cells. Biotechnol Lett 2014; 36:1203-7. [PMID: 24563309 DOI: 10.1007/s10529-014-1478-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/20/2014] [Indexed: 10/25/2022]
Abstract
IR-789, a novel near-infrared fluorescent probe, was designed, synthesized, and applied to living cells. The probe exhibited better response fluorescence characteristics than the only FDA-approved agent, indocyanine green. Cell experiments showed that the probe had high affinity and without apparent cytotoxicity. Fluorescent image experiments in living MCF-7 cells (human breast adenocarcinoma cell line) further demonstrated the potential applications of the probe in biological systems. The probe effectively prevented the influence of autofluorescence and native cellular species in biological systems. It also exhibited high sensitivity, good photostability, and excellent cell membrane permeability.
Collapse
|
42
|
Hussain T, Nguyen QT. Molecular imaging for cancer diagnosis and surgery. Adv Drug Deliv Rev 2014; 66:90-100. [PMID: 24064465 DOI: 10.1016/j.addr.2013.09.007] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/07/2013] [Accepted: 09/13/2013] [Indexed: 12/11/2022]
Abstract
Novel molecular imaging techniques have the potential to significantly enhance the diagnostic and therapeutic approaches for cancer treatment. For solid tumors in particular, novel molecular enhancers for imaging modalities such as US, CT, MRI and PET may facilitate earlier and more accurate diagnosis and staging which are prerequisites for successful surgical therapy. Enzymatically activatable "smart" molecular MRI probes seem particularly promising because of their potential to image tumors before and after surgical removal without re-administration of the probe to evaluate completeness of surgical resection. Furthermore, the use of "smart" MR probes as part of screening programs may enable detection of small tumors throughout the body in at-risk patient populations. Dual labeling of molecular MR probes with fluorescent dyes can add real time intraoperative guidance facilitating complete tumor resection and preservation of important structures. A truly theranostic approach with the further addition of therapeutic agents to the molecular probe for adjuvant therapy is conceivable for the future.
Collapse
|
43
|
Li Y, Rey-Dios R, Roberts DW, Valdés PA, Cohen-Gadol AA. Intraoperative fluorescence-guided resection of high-grade gliomas: a comparison of the present techniques and evolution of future strategies. World Neurosurg 2013; 82:175-85. [PMID: 23851210 DOI: 10.1016/j.wneu.2013.06.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 04/15/2013] [Accepted: 06/29/2013] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Fluorescence guidance has a demonstrated potential in maximizing the extent of high-grade glioma resection. Different fluorophores (fluorescent biomarkers), including 5-aminolevulinic acid (5-ALA) and fluorescein, have been examined with the use of several imaging techniques. Our goal was to review the state of this technology and discuss strategies for more widespread adoption. METHODS We performed a Medline search using the key words "fluorescence," "intraoperative fluorescence-guided resection," "intraoperative image-guided resection," and "brain glioma" for articles from 1960 until the present. This initial search revealed 267 articles. Each abstract and article was reviewed and the reference lists from select articles were further evaluated for relevance. A total of 64 articles included information about the role of fluorescence in resection of high-grade gliomas and therefore were selectively included for our analysis. RESULTS 5-ALA and fluorescein sodium have shown promise as fluorescent markers in detecting residual tumor intraoperatively. These techniques have demonstrated a significant increase in the extent of tumor resection. Regulatory barriers have limited the use of 5-ALA and technological challenges have restricted the use of fluorescein and its derivatives in the United States. Limitations to this technology currently exist, such as the fact that fluorescence at tumor margins is not always reliable for identification of tumor-brain interface. CONCLUSIONS These techniques are safe and effective for increasing gross total resection. The development of more tumor-specific fluorophores is needed to resolve problems with subjective interpretation of fluorescent signal at tumor margins. Techniques such as quantum dots and polymer or iron oxide-based nanoparticles have shown promise as potential future tools.
Collapse
Affiliation(s)
- Yiping Li
- Goodman Campbell Brain and Spine, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Roberto Rey-Dios
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - David W Roberts
- Section of Neurosurgery, Department of Surgery, Dartmouth Medical School, Lebanon, New Hampshire, USA; Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Pablo A Valdés
- Section of Neurosurgery, Department of Surgery, Dartmouth Medical School, Lebanon, New Hampshire, USA; Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Aaron A Cohen-Gadol
- Goodman Campbell Brain and Spine, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
44
|
Yadav N, Bhatia S, Ragheb J, Song Y, Romero A, Oh S, Lin WC. Evaluating and improving the quality of time-dependent, diffuse reflectance spectroscopic signals measured from in vivo brain during craniotomy. Med Eng Phys 2013; 35:1551-7. [PMID: 23665345 DOI: 10.1016/j.medengphy.2013.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 04/05/2013] [Accepted: 04/17/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Optical spectroscopy can be used to assess the pathophysiological characteristics of diseased and injured biological tissue in vivo in a non-destructive way. It is often used in conjunction with a contact optical probe for the purposes of operating and sensing in a sterile field. Since the probe is often held by the hand of an investigator during data acquisition, any hand instability can affect the quality of acquired data and, hence, degrade the accuracy of diagnosis. This study was designed to quantitatively characterize these artifacts, and then propose an effective engineering solution to remove them. METHODS Time-dependent diffuse reflectance spectra (Rd(λ,t)) were acquired from the normal cortex region of pediatric patients undergoing epilepsy surgery. They were acquired at a rate of 33 Hz, and their range was 400 and 900 nm. Two distinct ways of collecting data were tested: one with the fiber optical probe held by the surgeon's hand during data acquisition, and the other with the probe held by a specially designed probe holder. The probe holder was designed and constructed to minimize the variations in probe contact pressure and contact point for the full duration of any given investigation. Spectral data acquired using versus not using the probe holder were characterized and compared in the time, wavelength, and frequency domains, using both descriptive and inferential statistics. RESULTS Hand motion manifested as strong random variations in Rd(λ,t) which impacted temporal and frequency characteristics of Rd(λ,t). The percentage standard deviation %STD of Rd(λ,t) acquired without probe holder could be as high as 60%, and they are significantly higher than those with probe holder at all wavelengths. This difference is especially prominent between 400 and 600 nm. Rd(λ,t) acquired without the probe holder also processed a higher spectral power energy in the frequency domain than those with the probe holder. The correlation analysis revealed that the hand motions induced synchronistic variations in Rd(λ,t) between 600 and 800 nm, but this synchronicity is not obvious between 400 and 600 nm. CONCLUSION The results of this investigation demonstrate the nature and the magnitude of hand motion induced artifacts in in vivo diffuse reflectance spectra and propose one potential solution (i.e., a probe holder) to remove them. These findings allow us to improve the quality of time-dependent, diffuse reflectance signals acquired to study the dynamic characteristics of biological tissues, like brain, in vivo.
Collapse
Affiliation(s)
- Nitin Yadav
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Zhang H, Uselman RR, Yee D. Exogenous near-infrared fluorophores and their applications in cancer diagnosis: biological and clinical perspectives. ACTA ACUST UNITED AC 2013; 5:241-51. [PMID: 21566703 DOI: 10.1517/17530059.2011.566858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Near-infrared fluorescent (NIRF) imaging is a rapidly growing research field which has the potential to be an important imaging modality in cancer diagnosis. Various exogenous NIR fluorophores have been developed for the technique, including small molecule fluorophores and nanoparticles. NIRF imaging has been used in animal models for the detection of cancer overthe last twenty years and has in recent years been used in human clinical trials. AREAS COVERED This article describes the types and characteristics of exogenous fluorophores available for in vivo fluorescent cancer imaging. The article also discusses the progression of NIRF cancer imaging over recent years and its future challenges, from both a biological and clinical perspective. in The review also looks at its application for lymph node mapping, tumor targeting and characterization, and tumor margin definition for surgical guidance. EXPERT OPINION NIRF imaging is not in routine clinical cancer practice; yet, the authors predict that techniques using NIR fluorophores for tumor margin definition and lymph node mapping will enter clinical practice in the near future. The authors also anticipate that NIRF imaging research will lead to the development of flurophores with 'high brightness' that will overcome the limited penetration of this modality and be better suited for non invasive tumor targeting.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Medicine, Masonic Cancer Center, MMC 806, 420 Delaware St SE, Minneapolis, MN, 55455, USA,
| | | | | |
Collapse
|
46
|
Intraoperative fluorescent imaging of intracranial tumors: a review. Clin Neurol Neurosurg 2013; 115:517-28. [PMID: 23523009 DOI: 10.1016/j.clineuro.2013.02.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 02/15/2013] [Accepted: 02/18/2013] [Indexed: 11/24/2022]
Abstract
A review of fluorescent imaging for intracranial neoplasms is presented. Complete resection of brain cancer is seldom possible because of the goal to preserve brain tissue and the inability to visualize individual infiltrative tumor cells. Verification of histology and identification of tumor invasion in macroscopically normal-appearing brain tissue determine prognosis after resection of malignant gliomas. Therefore, imaging modalities aim to facilitate intraoperative decision-making. Intraoperative fluorescent imaging techniques have the potential to enable precise histopathologic diagnosis and to detect tumor remnants in the operative field. Macroscopic fluorescence imaging is effective for gross tumor detection. Microscopic imaging techniques enhance the sensitivity of the macroscopic observations and provide real-time histological information. Further development of clinical grade fluorescent agents specifically targeting tumor cells could improve the diagnostic and prognostic yield of intraoperative imaging.
Collapse
|
47
|
Abstract
Non-invasive optical imaging techniques, such as fluorescence imaging (FI) or bioluminescence imaging (BLI) have emerged as important tools in biomedical research. As demonstrated in different animal disease models, they enable visualization of physiological and pathophysiological processes at the cellular and molecular level in vivo with high specificity. Optical techniques are easy to use, fast, and affordable. Furthermore, they are characterized by their high sensitivity. In FI, very low amounts of the imaging agent (nano- to femtomol or even less) can be detected. Due to the absorption and scattering of light in tissue, optical techniques exhibit a comparably low spatial resolution in the millimeter range and a depth limit of a few centimeters. However, non-invasive imaging of biological processes in small animals and in outer or inner surfaces as well as during surgery even in humans is feasible. Currently two agents for fluorescence imaging are clinically approved, namely indocyanine green (ICG) and 5-aminolevulinic acid (5-ALA). In the past years, a number of new optical imaging agents for FI and reporter systems for BLI have been developed and successfully tested in animal models. Some of the FI agents might promise the application in clinical oncology. In this chapter, we describe the basic principles of non-invasive optical imaging techniques, give examples for the visualization of biological processes in animal models of cancer, and discuss potential clinical applications in oncology.
Collapse
|
48
|
Wyatt SK, Manning HC, Bai M, Ehtesham M, Mapara KY, Thompson RC, Bornhop DJ. Preclinical molecular imaging of the translocator protein (TSPO) in a metastases model based on breast cancer xenografts propagated in the murine brain. Curr Mol Med 2012; 12:458-66. [PMID: 22348613 DOI: 10.2174/156652412800163361] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/09/2012] [Accepted: 02/02/2012] [Indexed: 01/14/2023]
Abstract
Previous studies have demonstrated the feasibility of translocator protein (TSPO) imaging to visualize and quantify human breast adenocarcinoma (MDA-MB-231) cells in vivo using a TSPO-targeted near-infrared (NIR) probe (NIR-conPK11195). This study aimed to extend the use of the TSPO-targeted probe to a more biologically relevant and clinically important tumor microenvironment as well as to assess our ability to longitudinally detect the presence and progression of breast cancer cells in the brain. The in vivo biodistribution and accumulation of NIR-conPK11195 and free (unconjugated) NIR dye were quantitatively evaluated in intracranial MDA-MB-231-bearing mice and non-tumor-bearing control mice longitudinally once a week from two to five weeks post-inoculation. The in vivo time-activity curves illustrate distinct clearance profiles for NIR-conPK11195 and free NIR dye, resulting in preferential accumulation of the TSPO-targeted probe in the intracranial tumor bearing hemisphere (TBH) with significant tumor contrast over normal muscle tissue (p < 0.005 at five weeks; p < 0.01 at four weeks). In addition, the TSPO-labeled TBHs demonstrated significant contrast over the TBHs of mice injected with free NIR dye (p < 0.001 at four and five weeks) as well as over the TSPO-labeled non-tumor-bearing hemispheres (NTBHs) of control mice (p < 0.005 at four and five weeks). Overall, TSPO-targeted molecular imaging appears useful for visualizing and quantifying breast cancer xenografts propagated in the murine brain and may assist in preclinical detection, diagnosis and monitoring of metastatic disease as well as drug discovery. Furthermore, these results indicate it should be possible to perform TSPO-imaging of breast cancer cells in the brain using radiolabeled TSPO-targeted agents, particularly in light of the fact that [11C]-labeled TSPO probes such as [11C]-PK 11195 have been successfully used to image gliomas in the clinic.
Collapse
Affiliation(s)
- Shelby K Wyatt
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Meier R, Thürmel K, Moog P, Noël PB, Ahari C, Sievert M, Dorn F, Waldt S, Schaeffeler C, Golovko D, Haller B, Ganter C, Weckbach S, Woertler K, Rummeny EJ. Detection of synovitis in the hands of patients with rheumatologic disorders: Diagnostic performance of optical imaging in comparison with magnetic resonance imaging. ACTA ACUST UNITED AC 2012; 64:2489-98. [DOI: 10.1002/art.34467] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
50
|
Marcu L, Hartl BA. Fluorescence Lifetime Spectroscopy and Imaging in Neurosurgery. IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS : A PUBLICATION OF THE IEEE LASERS AND ELECTRO-OPTICS SOCIETY 2012; 18:1465-1477. [PMID: 28053498 PMCID: PMC5205025 DOI: 10.1109/jstqe.2012.2185823] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Clinical outcome of patients diagnosed with primary brain tumor has been correlated with the extent of surgical resection. In treating this disease, the neurosurgeon must balance between an aggressive, radical resection and minimizing the loss of healthy, functionally significant brain tissue. Numerous intra-operative methodologies and technological approaches have been explored as a means to improve the accuracy of surgical resection. This paper presents an overview of current conventional techniques and new emerging technologies with potential to impact the area of image-guided surgery of brain tumors. Emphasis is placed on techniques based on endogenous fluorescence lifetime contrast and their potential for intraoperative diagnosis of brain tumors.
Collapse
Affiliation(s)
- Laura Marcu
- University of California, Davis, Davis, CA 95616 USA
| | - Brad A Hartl
- University of California, Davis, Davis, CA 95616 USA
| |
Collapse
|