1
|
Romeo E, Tzakos AG, Crook T, Syed N, Voulgaris S, Alexiou GA. Agents for Fluorescence-Guided Glioblastoma Surgery. Pharmaceutics 2025; 17:637. [PMID: 40430928 PMCID: PMC12115318 DOI: 10.3390/pharmaceutics17050637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor, characterized by rapid progression and a median survival of no more than 12-18 months. Fluorescence-guided surgery is crucial, as it allows for tumor visualization and aids in its complete removal, which is essential for improving survival rates. We conducted a literature review to identify fluorescent agents that have been utilized in the removal of GBM and to assess their benefits in achieving maximum tumor resection. Our analysis focuses on their advantages, limitations, and potential impact on improving surgical precision and patient outcomes. We searched the PubMed database for studies published on fluorescence-guided resection of GBM and evaluated the utility of each agent in terms of outcomes, gross total resection (GTR), and their sensitivity and specificity for the tumor. The literature review revealed that the three agents successfully utilized are 5-aminolevulinic acid (5-ALA), sodium fluorescein, and indocyanine green. In addition to these, a variety of dyes have been investigated in studies, including peptides, lipids, and nanosystems, which appear to be very promising. To date, numerous fluorescent agents have been proposed for the surgical resection of GBM. However, 5-aminolevulinic acid (5-ALA) remains the only agent widely adopted in clinical practice, as its safety and efficacy have been well-established. Further clinical trials and studies are necessary to assess the utility, effectiveness, and potential advantages of emerging fluorescent dyes in enhancing GBM resection and improving patient outcomes.
Collapse
Affiliation(s)
- Eleni Romeo
- Neurosurgical Institute, University of Ioannina, 45500 Ioannina, Greece; (E.R.); (S.V.)
- Department of Neurosurgery, School of Medicine, University of Ioannina, 45500 Ioannina, Greece
| | - Andreas G. Tzakos
- Section of Organic Chemistry and Biochemistry, Department of Chemistry, University of Ioannina, 45500 Ioannina, Greece;
| | - Timothy Crook
- Department of Brain Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK; (T.C.); (N.S.)
| | - Nelofer Syed
- Department of Brain Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK; (T.C.); (N.S.)
| | - Spyridon Voulgaris
- Neurosurgical Institute, University of Ioannina, 45500 Ioannina, Greece; (E.R.); (S.V.)
- Department of Neurosurgery, School of Medicine, University of Ioannina, 45500 Ioannina, Greece
| | - George A. Alexiou
- Neurosurgical Institute, University of Ioannina, 45500 Ioannina, Greece; (E.R.); (S.V.)
- Department of Neurosurgery, School of Medicine, University of Ioannina, 45500 Ioannina, Greece
| |
Collapse
|
2
|
Lavrador JP, Wroe-Wright O, Marchi F, Elhag A, O’Keeffe A, De La Fuente P, Soumpasis C, Cardia A, Mirallave-Pescador A, Díaz-Baamonde A, Mosquera JS, Coiteiro D, Jewell S, Strong A, Gullan R, Ashkan K, Vergani F, Vasan AK, Bhangoo R. Microvascular Cortical Dynamics in Minimal Invasive Deep-Seated Brain Tumour Surgery. Cancers (Basel) 2025; 17:1392. [PMID: 40361321 PMCID: PMC12070978 DOI: 10.3390/cancers17091392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND The tubular retractor-assisted minimally invasive parafascicular approach (trMIPS) is a transsulcal approach to deep-seated brain tumours. It is a safe surgical approach but its impact on the microvascular dynamics of the retracted cortex and its clinical implications are unknown. METHODS This was a single-centre prospective study including patients with deep-seated brain tumours operated on with a trMIPS (BrainPath Nico System©). All patients underwent pre- and post-cannulation indocyanine green study using a FLOW 800 module in a KINEVO Zeiss© microscope. Speed, delay, time-to-peak (TtP) rise-in-time and cerebral blood flow index (CBFI) metrics were assessed. RESULTS Thirty-five patients were included, with 144 regions-of-interest (ROIs) selected. The majority of patients were diagnosed with glioblastoma (51.43%), and 37.14% of patients had a preoperative focal neurological deficit (FND) at presentation. A ROI-based analysis concluded that an increase in speed and CBFI was related with a worse neurological outcome when comparing the pre- and post-brain cannulation assessments (speed: deterioration = 43.12 ± 80.60% versus stable = -14.51 ± 57.80% versus improvement = 6.93 ± 31.33%, p < 0.0001; CBFI: deterioration = 50.40 ± 88.17% versus stable = -2.70 ± 67.54% versus improvement = -38.98 ± 26.17%, p = 0.0005). These findings were reproducible in a combined-ROI per patient analysis and confirmed after adjustment for confounding. CONCLUSION Microvascular flow dynamics impact trMIPS outcomes as an increase in the speed and CBFI after decannulation was related with worse neurological outcome.
Collapse
Affiliation(s)
- José Pedro Lavrador
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| | - Oliver Wroe-Wright
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| | - Francesco Marchi
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
- Department of Neurosurgery, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Lugano, Switzerland;
| | - Ali Elhag
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| | - Andrew O’Keeffe
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| | - Pablo De La Fuente
- Department of Neurosurgery, Araba University Hospital, 01009 Vitoria, Spain;
| | - Christos Soumpasis
- Department of Neurosurgery, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK;
| | - Andrea Cardia
- Department of Neurosurgery, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Lugano, Switzerland;
| | - Ana Mirallave-Pescador
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
- Department of Neurophysiology, King’s College Hospital Foundation Trust, London SE5 9RS, UK
| | - Alba Díaz-Baamonde
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
- Department of Neurophysiology, King’s College Hospital Foundation Trust, London SE5 9RS, UK
| | - Jose Sadio Mosquera
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
- Department of Neurophysiology, King’s College Hospital Foundation Trust, London SE5 9RS, UK
| | - Domingos Coiteiro
- Unidade de Saude Local, Hospital Santa Maria, 1649-028 Lisboa, Portugal;
| | - Sharon Jewell
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College University, London WC2R 2LS, UK; (S.J.); (A.S.)
| | - Anthony Strong
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College University, London WC2R 2LS, UK; (S.J.); (A.S.)
| | - Richard Gullan
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| | - Keyoumars Ashkan
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| | - Francesco Vergani
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| | - Ahilan Kailaya Vasan
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| | - Ranjeev Bhangoo
- Department of Neurosurgery, King’s College Hospital Foundation Trust, London SE5 9RS, UK; (J.P.L.); (O.W.-W.); (A.E.); (A.O.); (A.M.-P.); (A.D.-B.); (J.S.M.); (R.G.); (K.A.); (F.V.); (A.K.V.); (R.B.)
| |
Collapse
|
3
|
Roddan A, Czempiel T, Xu C, Xu H, Weld A, Chalau V, Anichini G, Elson DS, Giannarou S. Multimodal imaging platform for enhanced tumor resection in neurosurgery: integrating hyperspectral and pCLE technologies. Int J Comput Assist Radiol Surg 2025:10.1007/s11548-025-03340-1. [PMID: 40180672 DOI: 10.1007/s11548-025-03340-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE This work presents a novel multimodal imaging platform that integrates hyperspectral imaging (HSI) and probe-based confocal laser endomicroscopy (pCLE) for improved brain tumor identification during neurosurgery. By combining these two modalities, we aim to enhance surgical navigation, addressing the limitations of using each modality when used independently. METHODS We developed a multimodal imaging platform that integrates HSI and pCLE within an operating microscope setup using computer vision techniques. The system combines real-time, high-resolution HSI for macroscopic tissue analysis with pCLE for cellular-level imaging. The predictions of each modality made using Machine Learning methods are combined to improve tumor identification. RESULTS Our evaluation of the multimodal system revealed low spatial error, with minimal reprojection discrepancies, ensuring precise alignment between the HSI and pCLE. This combined imaging approach together with our multimodal tissue characterization algorithm significantly improves tumor identification, yielding higher Dice and Recall scores compared to using HSI or pCLE individually. CONCLUSION Our multimodal imaging platform represents a crucial first step toward enhancing tumor identification by combining HSI and pCLE modalities for the first time. We highlight improvements in metrics such as the Dice score and Recall, underscoring the potential for further advancements in this area.
Collapse
Affiliation(s)
- Alfie Roddan
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK.
| | - Tobias Czempiel
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Chi Xu
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Haozheng Xu
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Alistair Weld
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Vadzim Chalau
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Giulio Anichini
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Daniel S Elson
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| | - Stamatia Giannarou
- The Hamlyn Centre for Robotic Surgery, Imperial College London, Exhibition Rd, London, SW7 2AZ, UK
| |
Collapse
|
4
|
Xu Y, Zhang T, Li Z, Gao W, Guo K, Zhang Z, Zhang Z, Liu P. Fluorescence Endoscopy with Second Window Indocyanine Green for Surgical Resection of Malignant Brain Tumors. World Neurosurg 2025; 196:123766. [PMID: 39955047 DOI: 10.1016/j.wneu.2025.123766] [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: 01/22/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE This study aimed to validate the clinical feasibility, safety, and effectiveness of using the fluorescence endoscopy with second window indocyanine green (FESWICG) technique for resection of malignant brain tumors. METHODS Twenty-two patients undergoing surgery for malignant brain tumor resection were examined. Intravenous ICG (250 mg) was administered within 24 hours prior to surgery. All procedures were performed under endoscopic guidance. The fluorescence intensity of the tumors was monitored using fluorescence mode endoscopy. Specimens including resection margins were harvested and submitted for histopathological analysis. The sensitivity and specificity of FESWICG were validated using contrast-enhanced cranial magnetic resonance imaging performed within 24 hours of surgery and histopathology. The Karnofsky performance scale was used to assess overall functional status before and after surgery. RESULTS The tumor diagnoses were as follows: glioma (n = 14), ependymoma (n = 1), metastasis (n = 5), lymphoma (n = 1), and choroid plexus papilloma (n = 1). Intraoperative tumor fluorescence was strong in 20 patients and weak in 2. Postoperative contrast-enhanced imaging revealed complete tumor resection in 18 patients (81.82%). Sixty-four tumor specimens were collected, including 42 obtained from tumor margins. Using histopathology as the reference, the sensitivity, specificity, positive predictive value, and negative predictive value of FESWICG for detection of malignant brain tumors were 91.42%, 41.38%, 65.31%, and 80%, respectively. The median Karnofsky performance scale score was 85 before surgery and 93 at the 3-month follow-up. CONCLUSIONS SWICG notably enhanced intraoperative visualization of malignant brain tumors, particularly the delineation between tumor and normal brain. Its utility for margin detection is promising. When utilized in conjunction with a full endoscopic system, the visual acuity and overall effectiveness of surgical procedures can be substantially enhanced. However, ICG remains a low-specificity technique.
Collapse
Affiliation(s)
- Yongqiang Xu
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Tao Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Zhuoqun Li
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wenbo Gao
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ke Guo
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Zhao Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Zhirou Zhang
- Department of Rehabilitation Therapeutics, Binzhou Medical University, Yantai, Shandong, China
| | - Pengfei Liu
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
5
|
Shevtsov M, Yudintceva N, Bobkov D, Likhomanova R, Nechaeva A, Mikhailova E, Oganesyan E, Fedorov V, Kurkin A, Lukacheva A, Fofanov G, Kim A, Fedorov E, Sitovskaya D, Ulitin A, Mikhailova N, Anufriev I, Istomina M, Murashko E, Kessenikh E, Aksenov N, Vakhitova Y, Samochernykh K, Pitkin E, Shlyakhto E, Combs SE. RAS70 peptide targets multiforme glioblastoma by binding to the plasma membrane heat shock protein HSP70. Front Oncol 2025; 15:1543657. [PMID: 40196735 PMCID: PMC11973282 DOI: 10.3389/fonc.2025.1543657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 04/09/2025] Open
Abstract
Multiforme glioblastoma-homing peptides, particularly targeting plasma membrane-bound heat shock protein mHsp70, demonstrate great application potential for tumor theranostics. In the current study, to further increase the bioavailability as well as penetration capacity through the blood-brain barrier (BBB) of the mHsp70-targeted peptide TKDNNLLGRFELSG, which is known to bind to the oligomerization sequence of mHsp70 chaperone, the latter was conjugated with tripeptide RGD (forming chimeric peptide termed RAS70). In the model BBB system RAS70 efficiently crossed the barrier accumulating in the glioblastoma cells. Subsequently, in the orthotopic glioma models, intravenous administration of the fluorescently labeled agent (RAS70-sCy7.5) resulted in the tumor retention of peptide (further confirmed by histological studies). Thus, as shown by the biodistribution studies employing epifluorescence imaging, accumulation of RAS70-sCy7.5 in C6 glioma was significantly enhanced as compared to scramble peptide. Local application of the RAS70-sCy7.5 peptide that was sprayed over the dissected brain tissues helped to efficiently delineate the tumors in glioma-bearing animals employing an intraoperative fluorescent imaging system. Tumor-specific internalization of the peptide was further confirmed on the ex vivo primary GBM samples obtained from adult neurooncological patients. In conclusion, RAS70 peptide demonstrated high glioma-homing properties which could be employed for the intraoperative tumor visualization as well as for developing a potential carrier for drug delivery.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Natalia Yudintceva
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Danila Bobkov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
- Smorodintsev Research Institute of Influenza, St. Petersburg, Russia
| | - Ruslana Likhomanova
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Anastasiya Nechaeva
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Elena Mikhailova
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Elena Oganesyan
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Viacheslav Fedorov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Andrey Kurkin
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Anastasiya Lukacheva
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Georgii Fofanov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Aleksander Kim
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Evegeniy Fedorov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Daria Sitovskaya
- Polenov Neurosurgical Institute, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Alexey Ulitin
- Polenov Neurosurgical Institute, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Natalia Mikhailova
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Ilya Anufriev
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Maria Istomina
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Ekaterina Murashko
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Elizaveta Kessenikh
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Nikolay Aksenov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Yulia Vakhitova
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Konstantin Samochernykh
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
- Polenov Neurosurgical Institute, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Emil Pitkin
- Department of Statistics and Data Science, Wharton School, University of Pennsylvania, Philadelphia, PA, United States
| | - Evgeny Shlyakhto
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Stephanie E. Combs
- Department of Radiation Oncology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
6
|
Zamorano M, Zhu B, Massoud AT, Hendricks J, Choi HA, Peesh P, Miller BA, Zhang XR, Shah MN, Sevick-Muraca EM. Safety of Indocyanine Green Microdosing for Clinical Imaging of CSF Ventricular Dynamics and Extracranial Outflow. J Neuroimaging 2025; 35:e70028. [PMID: 40029024 DOI: 10.1111/jon.70028] [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: 12/14/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND AND PURPOSE Intravenous administration of indocyanine green (ICG) has been approved in brain surgeries for decades, yet concerns about neurotoxicity prevent its direct administration into the cerebrospinal fluid (CSF). Armed with prior animal studies demonstrating the feasibility of using ICG microdosing into the CSF, we sought to evaluate its nonclinical safety profile and obtain surrogate measures in adults prior to its use in human neonates. METHODS Evaluation of ICG toxicity was conducted in mixed primary CNS cell cultures and in an extended safety study of juvenile rat pups deploying intraventricular injections of saline (as control) or ICG. Analysis of animal behavior included Novel Object Place Recognition Test and rotarod behavioral tests. Immunohistochemical analysis of tumor necrosis factor-alpha (TNF-α), oxidative deoxyribonucleic acid damage, microglial activation, and neuronal density was performed on collected brains. We measured ICG levels (before and after intravenous administration) in collected CSF from external ventricular drain catheters of 10 brain-injured adults. RESULTS TNF-α and lactate dehydrogenase assay for cytotoxicity showed transient elevations after 1 h of incubation with 1291 µM ICG, but none at or below 322 µM ICG, even after 24 h of incubation. Behavioral tests and immunohistochemical analyses showed no differences between ICG-administered animals and controls. Intraventricular concentrations of ICG in collected human CSF ranged between 0.17 and 7.93 µM, with no adverse events associated with intravenous administration. CONCLUSIONS With intraventricular microdosing of 100 µg ICG, maximal ICG concentrations in neonatal CSF range from 1.3 to 6 5 µM. CNS cell culture, rat safety studies, and surrogate measures in adults evidence the safety of microdosing ICG directly into the CSF.
Collapse
Affiliation(s)
- Miriam Zamorano
- Department of Pediatric Surgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - Banghe Zhu
- Department of Pediatric Surgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - Ahmed T Massoud
- Department of Pediatric Surgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
- Department of Neurosurgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - Jonathan Hendricks
- Department of Pediatric Surgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - H Alex Choi
- Department of Neurosurgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - Pedram Peesh
- Department of Neurosurgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - Brandon A Miller
- Department of Pediatric Surgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
- Department of Neurosurgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - Xinhai Robert Zhang
- Department of Pathology, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - Manish N Shah
- Department of Pediatric Surgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
- Department of Neurosurgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| | - Eva M Sevick-Muraca
- Department of Pediatric Surgery, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, McGovern School of Medicine, The University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
7
|
Wårdell K, Klint E, Richter J. Probe-Based Fluorescence Spectroscopy for In Situ Brain Tumor Measurements During Resection and Needle Biopsies. Biomedicines 2025; 13:537. [PMID: 40149515 PMCID: PMC11940453 DOI: 10.3390/biomedicines13030537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Primary brain tumors are difficult to identify intraoperatively due to their infiltrative character in the marginal zone. Several optical methods have been suggested. Of these, 5-ALA-induced fluorescence visualized through a microscope is the most common. The aim is to present an investigational probe-based optical system and its translation for clinical use, summarize previous studies, and give examples of clinical implementations during resection and burr hole biopsies. Methods: The FluoRa system combines 5-ALA fluorescence spectroscopy with laser Doppler flowmetry (LDF). Probe designs are available for brain tumor resection (hand-held probe) or burr hole needle biopsies (frame-based or navigated). The outer cannulas of biopsy needles are modified with an opening at the tip for simultaneous use with optical probes during insertion along the trajectory. An updated version of FluoRa is introduced and experimentally investigated. Results: Probe-based fluorescence spectroscopy has been successfully translated for clinical use and applied during brain tumor resection (n = 75) and burr hole needle biopsies (n = 47). Forward-looking optical measurements through the biopsy needle reduce the number of trajectories (28/27) compared to prior to insertion (28/20), at the same time that the target for tissue sampling can be identified in situ. Additionally, increased microcirculation is identified along the trajectory with LDF. This is accomplished with FluoRa. Conclusions: Intraoperative probe-based spectroscopic measurements quantify 5-ALA fluorescence and thus identify glioblastoma and lymphoma tissue in situ during resection and burr hole needle biopsies.
Collapse
Affiliation(s)
- Karin Wårdell
- Department of Biomedical Engineering, Linköping University, 581 85 Linköping, Sweden; (E.K.); (J.R.)
| | - Elisabeth Klint
- Department of Biomedical Engineering, Linköping University, 581 85 Linköping, Sweden; (E.K.); (J.R.)
| | - Johan Richter
- Department of Biomedical Engineering, Linköping University, 581 85 Linköping, Sweden; (E.K.); (J.R.)
- Department of Neurosurgery, Linköping University Hospital, 581 85 Linköping, Sweden
| |
Collapse
|
8
|
Cipolato O, Fauconneau M, LeValley PJ, Nißler R, Suter B, Herrmann IK. An Augmented Reality Visor for Intraoperative Visualization, Guidance, and Temperature Monitoring Using Fluorescence. JOURNAL OF BIOPHOTONICS 2025; 18:e202400417. [PMID: 39716019 PMCID: PMC11793943 DOI: 10.1002/jbio.202400417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
Fluorescence-guided surgeries, including tumor resection and tissue soldering, are advancing the frontiers of surgical precision by offering enhanced control that minimizes tissue damage, improving recovery and outcomes. However, integrating fluorescence visualization with real-time temperature monitoring remains a challenge, limiting broader clinical use. We address this issue with an augmented reality (AR) visor that combines nanomaterial excitation, fluorescence detection, and temperature monitoring. Using advanced fluorescent nanoparticles like indocyanine green-doped particles and carbon nanotubes, the visor provides a comprehensive view of both the surgical field and sub-surface conditions invisible to the naked eye. This integration improves the safety and efficacy of fluorescence-guided surgeries, including laser tissue soldering, by ensuring optimal temperatures and laser guidance in real time. The presented technology enhances existing surgical techniques and supports the development of new strategies and sensing technologies in areas where traditional methods fall short, marking significant progress in precision surgery and potentially improving patient care.
Collapse
Affiliation(s)
- Oscar Cipolato
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering (D‐MAVT), Institute of Energy and Process Engineering (IEPE)ETH ZurichZurichSwitzerland
- Particles Biology Interactions Laboratory, Department of Materials Meet LifeSwiss Federal Laboratories for Materials Science and Technology (Empa)St. GallenSwitzerland
- The Ingenuity LabUniversity Hospital BalgristZurichSwitzerland
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
| | - Matthias Fauconneau
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering (D‐MAVT), Institute of Energy and Process Engineering (IEPE)ETH ZurichZurichSwitzerland
| | - Paige J. LeValley
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering (D‐MAVT), Institute of Energy and Process Engineering (IEPE)ETH ZurichZurichSwitzerland
- Particles Biology Interactions Laboratory, Department of Materials Meet LifeSwiss Federal Laboratories for Materials Science and Technology (Empa)St. GallenSwitzerland
- The Ingenuity LabUniversity Hospital BalgristZurichSwitzerland
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
| | - Robert Nißler
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering (D‐MAVT), Institute of Energy and Process Engineering (IEPE)ETH ZurichZurichSwitzerland
- Particles Biology Interactions Laboratory, Department of Materials Meet LifeSwiss Federal Laboratories for Materials Science and Technology (Empa)St. GallenSwitzerland
- The Ingenuity LabUniversity Hospital BalgristZurichSwitzerland
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
| | - Benjamin Suter
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering (D‐MAVT), Institute of Energy and Process Engineering (IEPE)ETH ZurichZurichSwitzerland
- Particles Biology Interactions Laboratory, Department of Materials Meet LifeSwiss Federal Laboratories for Materials Science and Technology (Empa)St. GallenSwitzerland
- The Ingenuity LabUniversity Hospital BalgristZurichSwitzerland
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
| | - Inge K. Herrmann
- Nanoparticle Systems Engineering Laboratory, Department of Mechanical and Process Engineering (D‐MAVT), Institute of Energy and Process Engineering (IEPE)ETH ZurichZurichSwitzerland
- Particles Biology Interactions Laboratory, Department of Materials Meet LifeSwiss Federal Laboratories for Materials Science and Technology (Empa)St. GallenSwitzerland
- The Ingenuity LabUniversity Hospital BalgristZurichSwitzerland
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
| |
Collapse
|
9
|
Kotwal A, Saragadam V, Bernstock JD, Sandoval A, Veeraraghavan A, Valdés PA. Hyperspectral imaging in neurosurgery: a review of systems, computational methods, and clinical applications. JOURNAL OF BIOMEDICAL OPTICS 2025; 30:023512. [PMID: 39544341 PMCID: PMC11559659 DOI: 10.1117/1.jbo.30.2.023512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 11/17/2024]
Abstract
Significance Accurate identification between pathologic (e.g., tumors) and healthy brain tissue is a critical need in neurosurgery. However, conventional surgical adjuncts have significant limitations toward achieving this goal (e.g., image guidance based on pre-operative imaging becomes inaccurate up to 3 cm as surgery proceeds). Hyperspectral imaging (HSI) has emerged as a potential powerful surgical adjunct to enable surgeons to accurately distinguish pathologic from normal tissues. Aim We review HSI techniques in neurosurgery; categorize, explain, and summarize their technical and clinical details; and present some promising directions for future work. Approach We performed a literature search on HSI methods in neurosurgery focusing on their hardware and implementation details; classification, estimation, and band selection methods; publicly available labeled and unlabeled data; image processing and augmented reality visualization systems; and clinical study conclusions. Results We present a detailed review of HSI results in neurosurgery with a discussion of over 25 imaging systems, 45 clinical studies, and 60 computational methods. We first provide a short overview of HSI and the main branches of neurosurgery. Then, we describe in detail the imaging systems, computational methods, and clinical results for HSI using reflectance or fluorescence. Clinical implementations of HSI yield promising results in estimating perfusion and mapping brain function, classifying tumors and healthy tissues (e.g., in fluorescence-guided tumor surgery, detecting infiltrating margins not visible with conventional systems), and detecting epileptogenic regions. Finally, we discuss the advantages and disadvantages of HSI approaches and interesting research directions as a means to encourage future development. Conclusions We describe a number of HSI applications across every major branch of neurosurgery. We believe these results demonstrate the potential of HSI as a powerful neurosurgical adjunct as more work continues to enable rapid acquisition with smaller footprints, greater spectral and spatial resolutions, and improved detection.
Collapse
Affiliation(s)
- Alankar Kotwal
- University of Texas Medical Branch, Department of Neurosurgery, Galveston, Texas, United States
- Rice University, Department of Electrical and Computer Engineering, Houston, Texas, United States
| | - Vishwanath Saragadam
- University of California Riverside, Department of Electrical and Computer Engineering, Riverside, California, United States
| | - Joshua D. Bernstock
- Brigham and Women’s Hospital, Harvard Medical School, Department of Neurosurgery, Boston, Massachusetts, United States
- Massachusetts Institute of Technology, David H. Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts, United States
| | - Alfredo Sandoval
- University of Texas Medical Branch, Department of Neurosurgery, Galveston, Texas, United States
| | - Ashok Veeraraghavan
- Rice University, Department of Electrical and Computer Engineering, Houston, Texas, United States
| | - Pablo A. Valdés
- University of Texas Medical Branch, Department of Neurosurgery, Galveston, Texas, United States
- Rice University, Department of Electrical and Computer Engineering, Houston, Texas, United States
| |
Collapse
|
10
|
Scorzo AV, Kwon CY, Strawbridge RR, Duke RB, Chen KL, Li C, Fan X, Hoopes PJ, Roberts DW, Paulsen KD, Davis SC. Comparing spatial distributions of ALA-PpIX and indocyanine green in a whole pig brain glioma model using 3D fluorescence cryotomography. JOURNAL OF BIOMEDICAL OPTICS 2025; 30:S13704. [PMID: 39247519 PMCID: PMC11379406 DOI: 10.1117/1.jbo.30.s1.s13704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024]
Abstract
Significance ALA-PpIX and second-window indocyanine green (ICG) have been studied widely for guiding the resection of high-grade gliomas. These agents have different mechanisms of action and uptake characteristics, which can affect their performance as surgical guidance agents. Elucidating these differences in animal models that approach the size and anatomy of the human brain would help guide the use of these agents. Herein, we report on the use of a new pig glioma model and fluorescence cryotomography to evaluate the 3D distributions of both agents throughout the whole brain. Aim We aim to assess and compare the 3D spatial distributions of ALA-PpIX and second-window ICG in a glioma-bearing pig brain using fluorescence cryotomography. Approach A glioma was induced in the brain of a transgenic Oncopig via adeno-associated virus delivery of Cre-recombinase plasmids. After tumor induction, the pro-drug 5-ALA and ICG were administered to the animal 3 and 24 h prior to brain harvest, respectively. The harvested brain was imaged using fluorescence cryotomography. The fluorescence distributions of both agents were evaluated in 3D in the whole brain using various spatial distribution and contrast performance metrics. Results Significant differences in the spatial distributions of both agents were observed. Indocyanine green accumulated within the tumor core, whereas ALA-PpIX appeared more toward the tumor periphery. Both ALA-PpIX and second-window ICG provided elevated tumor-to-background contrast (13 and 23, respectively). Conclusions This study is the first to demonstrate the use of a new glioma model and large-specimen fluorescence cryotomography to evaluate and compare imaging agent distribution at high resolution in 3D.
Collapse
Affiliation(s)
- Augustino V. Scorzo
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Caleb Y. Kwon
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | | | - Ryan B. Duke
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Kristen L. Chen
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Chengpei Li
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Xiaoyao Fan
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - P. Jack Hoopes
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States
| | - David W. Roberts
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States
| | - Keith D. Paulsen
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States
| | - Scott C. Davis
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| |
Collapse
|
11
|
Mansour HM, Shah S, Aguilar TM, Abdul-Muqsith M, Gonzales-Portillo GS, Mehta AI. Enhancing Glioblastoma Resection with NIR Fluorescence Imaging: A Systematic Review. Cancers (Basel) 2024; 16:3984. [PMID: 39682171 DOI: 10.3390/cancers16233984] [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: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Glioblastoma (GB) is among the most aggressive and difficult-to-treat brain tumors, with a median survival of only 12-15 months despite maximal treatments, including surgery, radiotherapy, and chemotherapy. Extensive surgical resection improves survival in glioblastoma patients; however, achieving complete resection is often hindered by limitations in neurosurgical guidance technologies for accurate tumor margin detection. Recent advancements in fluorescence-guided surgery (FGS) and imaging techniques have significantly enhanced the precision and extent of glioblastoma resections. This study evaluates the impact of NIR fluorescence imaging on tumor visualization, surgical precision, cost-effectiveness, and patient survival. A systematic review of PubMed, Scopus, Google Scholar, and Embase was conducted to identify studies on the role of NIR fluorescence in glioblastoma surgery. A total of 135 studies were included, comprising 10 reviews, three clinical studies, 10 randomized controlled trials (RCTs), 10 preclinical studies, and four case reports, all focused on NIR fluorescence imaging in glioblastoma surgery. The findings indicate that NIR fluorescence imaging significantly improves tumor visualization, resulting in an 18-22% increase in gross total resection (GTR) rates in clinical studies. NIR fluorescence provides continuous real-time feedback, minimizing repeat imaging, reducing operational costs, and increasing GTR. These improvements contribute to better patient outcomes, including extended progression-free survival, improved overall survival, and reduced postoperative neurological deficits. This review underscores the potential of NIR imaging to establish a new standard for intraoperative glioblastoma management.
Collapse
Affiliation(s)
- Hadeel M Mansour
- Department of Neurosurgery, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| | - Tania M Aguilar
- Department of Neurosurgery, University of Illinois Chicago, Chicago, IL 60612, USA
| | | | | | - Ankit I Mehta
- Department of Neurosurgery, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
12
|
Körner LI, Reichert D, Andreana M, Unterhuber A, Erkkilae MT, Makolli J, Kiesel B, Mischkulnig M, Rötzer-Pejrimovsky T, Wöhrer A, Berger MS, Leitgeb R, Widhalm G. Analysis of the Porphyrin Peak Shift and Fluorescence Lifetime in Gliomas with Different Tumor Grades, Intratumoral Regions, and Visible Fluorescence Status. Diagnostics (Basel) 2024; 14:2651. [PMID: 39682559 DOI: 10.3390/diagnostics14232651] [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: 10/17/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Background: 5-aminolevulinic acid (5-ALA)-induced protoporphyrin IX (PpIX) fluorescence shows high sensitivity in detecting the tumor core of high-grade gliomas (HGG) but poor sensitivity for tissue of low-grade gliomas (LGG) and the margins of HGG. The characteristic emission peak for PpIX is known to be located at 635 nm. Recently, a second emission peak was described at 620 nm wavelength in LGG and the tumor infiltration zone of HGG. Methods: During surgery, samples from the tumor core and tumor infiltration zone of 43 WHO grade 2-4 gliomas were collected after preoperative 5-ALA administration, and their PpIX emission spectra, as well as fluorescence lifetimes, were determined by ex vivo analysis. Subsequently, the relative PpIX peak contribution (RPPC) was retrieved by calculating the integral of the two bands corresponding to the two emission peaks of PpIX (615-625 nm, 625-635 nm) and correlated with fluorescence lifetimes. Results: The mean RPPC decreased in samples with descending order of WHO grades, non-fluorescing samples, and infiltrative tumor regions, indicating a shift toward the 620 nm peak in porphyrin fluorescence. The porphyrin peak shift across all specimens correlated with lower fluorescence lifetimes (R: 0.854, R-squared: 0.729). Conclusions: The observed peak shift has important implications for fluorescence lifetime analyses since the lifetimes of other porphyrins contribute to the overall decay dynamics. Based on these initial data using fluorescence lifetime, this knowledge is of major importance, especially for detecting tissue from LGG that lack visible fluorescence, to further optimize the visualization of these tumor tissue using this promising imaging modality.
Collapse
Affiliation(s)
- Lisa Irina Körner
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, 1090 Vienna, Austria
| | - David Reichert
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
- Christian Doppler Laboratory OPTRAMED, Medical University of Vienna, 1090 Vienna, Austria
| | - Marco Andreana
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
| | - Angelika Unterhuber
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
| | - Mikael T Erkkilae
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
| | - Jessica Makolli
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Barbara Kiesel
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Mario Mischkulnig
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Rötzer-Pejrimovsky
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Adelheid Wöhrer
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Rainer Leitgeb
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
- Christian Doppler Laboratory OPTRAMED, Medical University of Vienna, 1090 Vienna, Austria
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
13
|
Aiudi D, Iacoangeli A, Mattioli A, Raggi A, Dobran M, Polonara G, Gigli R, Iacoangeli M, Gladi M. Cerebral Aneurysms and Arteriovenous Malformation: Preliminary Experience with the Use of Near-Infrared Fluorescence Imaging Applied to Endoscopy. J Pers Med 2024; 14:1117. [PMID: 39728030 DOI: 10.3390/jpm14121117] [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: 10/17/2024] [Revised: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Indocyanine green video angiography, integrated into the operative microscope, is frequently used in cerebrovascular surgery. This technology is often preferred, for cost or availability, to Doppler or intraoperative DSA (digital subtraction angiography). With the same assumption it was possible, in our preliminary experience, to partially vicariate the aforementioned devices using the SPY mode of the Stryker endoscope; it allowed the visualization of fluorescence in high definition. Methods: A retrospective analysis was conducted on a series of five patients suffering from cerebral aneurysm or AVM (arteriovenous malformation) who underwent, during the last year, surgical treatment with the aid of the microscope supported by the Stryker endoscope in the SPY mode for the visualization of the fluorescence emitted by indocyanine green. Results: All aneurysms were completely excluded from the cerebrovascular circulation in the absence of residues in the collar and occlusion of adjacent vessels; the complete removal of the nidus in all the AVMs was achieved with no residues. Conclusions: The intraoperative use of indocyanine green was a safe, rapid, and effective technique within a preliminary case study of "regular-not giant" aneurysms and superficially located AVM. The endoscopic technique in the SPY mode has allowed to partially vicariate the use of Doppler, intraoperative angiography, and integrated microscope video angiography. For these purposes, we propose, in selected cases, the support of the endoscope in the SPY mode during the microsurgical procedure in order to visualize the green fluorescence of indocyanine.
Collapse
Affiliation(s)
- Denis Aiudi
- Department of Neurosurgery, Marche Polytechnic University, 60126 Ancona, Italy
| | - Alessio Iacoangeli
- Department of Neurosurgery, Marche Polytechnic University, 60126 Ancona, Italy
| | - Andrea Mattioli
- Department of Neurosurgery, Marche Polytechnic University, 60126 Ancona, Italy
| | - Alessio Raggi
- Department of Neurosurgery, Marche Polytechnic University, 60126 Ancona, Italy
| | - Mauro Dobran
- Department of Neurosurgery, Marche Polytechnic University, 60126 Ancona, Italy
| | - Gabriele Polonara
- Department of Neuroradiology, Marche Polytechnic University, 60126 Ancona, Italy
| | - Riccardo Gigli
- Department of Neuroradiology, Marche Polytechnic University, 60126 Ancona, Italy
| | - Maurizio Iacoangeli
- Department of Neurosurgery, Marche Polytechnic University, 60126 Ancona, Italy
- IRCCS INRCA, Marche Polytechnic University, 60124 Ancona, Italy
| | - Maurizio Gladi
- Department of Neurosurgery, Marche Polytechnic University, 60126 Ancona, Italy
| |
Collapse
|
14
|
Khan M, Nasim M, Feizy M, Parveen R, Gull A, Khan S, Ali J. Contemporary strategies in glioblastoma therapy: Recent developments and innovations. Neuroscience 2024; 560:211-237. [PMID: 39368608 DOI: 10.1016/j.neuroscience.2024.09.022] [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: 07/11/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma multiforme (GBM) represents one of the most prevailing and aggressive primary brain tumors among adults. Despite advances in therapeutic approaches, the complex microenvironment of GBM poses significant challenges in its optimal therapy, which are attributed to immune evasion, tumor repopulation by stem cells, and limited drug penetration across the blood-brain barrier (BBB). Nanotechnology has emerged as a promising avenue for GBM treatment, offering biosafety, sustained drug release, enhanced solubility, and improved BBB penetrability. In this review, a comprehensive overview of recent advancements in nanocarrier-based drug delivery systems for GBM therapy is emphasized. The conventional and novel treatment modalities for GBM and the potential of nanocarriers to overcome existing limitations are comprehensively covered. Furthermore, the updates in the clinical landscape of GBM therapeutics are presented in addition to the current status of drugs and patents in the same context. Through a critical evaluation of existing literature, the therapeutic prospect and limitations of nanocarrier-based drug delivery strategies are highlighted offering insights into future research directions and clinical translation.
Collapse
Affiliation(s)
- Mariya Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Modassir Nasim
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Mohammadamin Feizy
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Rabea Parveen
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Azka Gull
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Saba Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India.
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India.
| |
Collapse
|
15
|
Zamay G, Koshmanova A, Narodov A, Gorbushin A, Voronkovskii I, Grek D, Luzan N, Kolovskaya O, Shchugoreva I, Artyushenko P, Glazyrin Y, Fedotovskaya V, Kuziakova O, Veprintsev D, Belugin K, Lukyanenko K, Nikolaeva E, Kirichenko A, Lapin I, Khorzhevskii V, Semichev E, Mohov A, Kirichenko D, Tokarev N, Chanchikova N, Krat A, Zukov R, Bakhtina V, Shnyakin P, Shesternya P, Tomilin F, Kosinova A, Svetlichnyi V, Zamay T, Kumeiko V, Mezko V, Berezovski MV, Kichkailo A. Visualization of Brain Tumors with Infrared-Labeled Aptamers for Fluorescence-Guided Surgery. J Am Chem Soc 2024; 146:24989-25004. [PMID: 39186481 PMCID: PMC11404482 DOI: 10.1021/jacs.4c06716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Gliomas remain challenging brain tumors to treat due to their infiltrative nature. Accurately identifying tumor boundaries during surgery is crucial for successful resection. This study introduces an innovative intraoperative visualization method utilizing surgical fluorescence microscopy to precisely locate tumor cell dissemination. Here, the focus is on the development of a novel contrasting agent (IR-Glint) for intraoperative visualization of human glial tumors comprising infrared-labeled Glint aptamers. The specificity of IR-Glint is assessed using flow cytometry and microscopy on primary cell cultures. In vivo effectiveness is studied on mouse and rabbit models, employing orthotopic xenotransplantation of human brain gliomas with various imaging techniques, including PET/CT, in vivo fluorescence visualization, confocal laser scanning, and surgical microscopy. The experiments validate the potential of IR-Glint for the intraoperative visualization of gliomas using infrared imaging. IR-Glint penetrates the blood-brain barrier and can be used for both intravenous and surface applications, allowing clear visualization of the tumor. The surface application directly to the brain reduces the dosage required and mitigates potential toxic effects on the patient. The research shows the potential of infrared dye-labeled aptamers for accurately visualizing glial tumors during brain surgery. This novel aptamer-assisted fluorescence-guided surgery (AptaFGS) may pave the way for future advancements in the field of neurosurgery.
Collapse
Affiliation(s)
- Galina Zamay
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Aptamerlab LLC, Krasnoyarsk 660042, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Anastasia Koshmanova
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Andrey Narodov
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Aptamerlab LLC, Krasnoyarsk 660042, Russia
| | - Anton Gorbushin
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Aptamerlab LLC, Krasnoyarsk 660042, Russia
- Krasnoyarsk Inter-District Ambulance Hospital Named after N.S. Karpovich, 17 Kurchatova, Krasnoyarsk 660062, Russia
| | - Ivan Voronkovskii
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Aptamerlab LLC, Krasnoyarsk 660042, Russia
- Krasnoyarsk Inter-District Ambulance Hospital Named after N.S. Karpovich, 17 Kurchatova, Krasnoyarsk 660062, Russia
| | - Daniil Grek
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Aptamerlab LLC, Krasnoyarsk 660042, Russia
| | - Natalia Luzan
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Olga Kolovskaya
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Aptamerlab LLC, Krasnoyarsk 660042, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Irina Shchugoreva
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Polina Artyushenko
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Yury Glazyrin
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Victoriya Fedotovskaya
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Olga Kuziakova
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Dmitry Veprintsev
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Kirill Belugin
- Federal Siberian Research Clinical Centre under the Federal Medical Biological Agency, Krasnoyarsk 660130, Russia
| | - Kirill Lukyanenko
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Elena Nikolaeva
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Andrey Kirichenko
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Aptamerlab LLC, Krasnoyarsk 660042, Russia
| | - Ivan Lapin
- Laboratory of Advanced Materials and Technology, Tomsk State University, Tomsk 634050, Russia
| | - Vladimir Khorzhevskii
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Krasnoyarsk Regional Pathology-Anatomic Bureau, Partizana Zheleznyaka, Krasnoyarsk 660022, Russia
| | - Evgeniy Semichev
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Alexey Mohov
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Daria Kirichenko
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Nikolay Tokarev
- Federal Siberian Research Clinical Centre under the Federal Medical Biological Agency, Krasnoyarsk 660130, Russia
| | - Natalia Chanchikova
- Federal Siberian Research Clinical Centre under the Federal Medical Biological Agency, Krasnoyarsk 660130, Russia
| | - Alexey Krat
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Krasnoyarsk Regional Clinical Cancer Center, 16 1-ya Smolenskaya, Krasnoyarsk 660133, Russia
| | - Ruslan Zukov
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Krasnoyarsk Regional Clinical Cancer Center, 16 1-ya Smolenskaya, Krasnoyarsk 660133, Russia
| | - Varvara Bakhtina
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Pavel Shnyakin
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Pavel Shesternya
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
| | - Felix Tomilin
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
- Kirensky Institute of Physics, Krasnoyarsk 660036, Russia
| | - Aleksandra Kosinova
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Valery Svetlichnyi
- Laboratory of Advanced Materials and Technology, Tomsk State University, Tomsk 634050, Russia
| | - Tatiana Zamay
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| | - Vadim Kumeiko
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok 690041, Russia
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | | | - Maxim V Berezovski
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Anna Kichkailo
- Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk 660022, Russia
- Aptamerlab LLC, Krasnoyarsk 660042, Russia
- Federal Research Center "Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences", Krasnoyarsk 660036, Russia
| |
Collapse
|
16
|
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
|
17
|
Chen H, Mirg S, Gaddale P, Agrawal S, Li M, Nguyen V, Xu T, Li Q, Liu J, Tu W, Liu X, Drew PJ, Zhang N, Gluckman BJ, Kothapalli S. Multiparametric Brain Hemodynamics Imaging Using a Combined Ultrafast Ultrasound and Photoacoustic System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401467. [PMID: 38884161 PMCID: PMC11336909 DOI: 10.1002/advs.202401467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/25/2024] [Indexed: 06/18/2024]
Abstract
Studying brain-wide hemodynamic responses to different stimuli at high spatiotemporal resolutions can help gain new insights into the mechanisms of neuro- diseases and -disorders. Nonetheless, this task is challenging, primarily due to the complexity of neurovascular coupling, which encompasses interdependent hemodynamic parameters including cerebral blood volume (CBV), cerebral blood flow (CBF), and cerebral oxygen saturation (SO2). The current brain imaging technologies exhibit inherent limitations in resolution, sensitivity, and imaging depth, restricting their capacity to comprehensively capture the intricacies of cerebral functions. To address this, a multimodal functional ultrasound and photoacoustic (fUSPA) imaging platform is reported, which integrates ultrafast ultrasound and multispectral photoacoustic imaging methods in a compact head-mountable device, to quantitatively map individual dynamics of CBV, CBF, and SO2 as well as contrast agent enhanced brain imaging at high spatiotemporal resolutions. Following systematic characterization, the fUSPA system is applied to study brain-wide cerebrovascular reactivity (CVR) at single-vessel resolution via relative changes in CBV, CBF, and SO2 in response to hypercapnia stimulation. These results show that cortical veins and arteries exhibit differences in CVR in the stimulated state and consistent anti-correlation in CBV oscillations during the resting state, demonstrating the multiparametric fUSPA system's unique capabilities in investigating complex mechanisms of brain functions.
Collapse
Affiliation(s)
- Haoyang Chen
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Shubham Mirg
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Prameth Gaddale
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Sumit Agrawal
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Menghan Li
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Van Nguyen
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Tianbao Xu
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Qiong Li
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Jinyun Liu
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Wenyu Tu
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Xiao Liu
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Institute for Computational and Data SciencesThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Patrick J. Drew
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of Engineering Science and MechanicsThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of BiologyThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of NeurosurgeryThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Nanyin Zhang
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Bruce J. Gluckman
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of Engineering Science and MechanicsThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of NeurosurgeryThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Sri‐Rajasekhar Kothapalli
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Center for Neural EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Penn State Cancer InstituteThe Pennsylvania State UniversityHersheyPA17033USA
- Graduate Program in AcousticsThe Pennsylvania State UniversityUniversity ParkPA16802USA
| |
Collapse
|
18
|
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
|
19
|
Sindeeva OA, Demina PA, Kozyreva ZV, Terentyeva DA, Gusliakova OI, Muslimov AR, Sukhorukov GB. Single Mesenchymal Stromal Cell Migration Tracking into Glioblastoma Using Photoconvertible Vesicles. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1215. [PMID: 39057891 PMCID: PMC11279842 DOI: 10.3390/nano14141215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Reliable cell labeling and tracking techniques are imperative for elucidating the intricate and ambiguous interactions between mesenchymal stromal cells (MSCs) and tumors. Here, we explore fluorescent photoconvertible nanoengineered vesicles to study mMSC migration in brain tumors. These 3 μm sized vesicles made of carbon nanoparticles, Rhodamine B (RhB), and polyelectrolytes are readily internalized by cells. The dye undergoes photoconversion under 561 nm laser exposure with a fluorescence blue shift upon demand. The optimal laser irradiation duration for photoconversion was 0.4 ms, which provided a maximal blue shift of the fluorescent signal label without excessive laser exposure on cells. Vesicles modified with an extra polymer layer demonstrated enhanced intracellular uptake without remarkable effects on cell viability, motility, or proliferation. The optimal ratio of 20 vesicles per mMSC was determined. Moreover, the migration of individual mMSCs within 2D and 3D glioblastoma cell (EPNT-5) colonies over 2 days and in vivo tumor settings over 7 days were traced. Our study provides a robust nanocomposite platform for investigating MSC-tumor dynamics and offers insights into envisaged therapeutic strategies. Photoconvertible vesicles also present an indispensable tool for studying complex fundamental processes of cell-cell interactions for a wide range of problems in biomedicine.
Collapse
Affiliation(s)
- Olga A. Sindeeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
| | - Polina A. Demina
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia;
| | - Zhanna V. Kozyreva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
| | - Daria A. Terentyeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
| | - Olga I. Gusliakova
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
- Science Medical Center, Saratov State University, 83 Astrakhanskaya Str., 410012 Saratov, Russia;
| | - Albert R. Muslimov
- Center for Molecular and Cell Technologies, Saint Petersburg State Chemical and Pharmaceutical University, 14 Professora Popova Str., lit. A, 197022 St. Petersburg, Russia;
| | - Gleb B. Sukhorukov
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skoltech, 3 Nobel Str., 121205 Moscow, Russia; (Z.V.K.); (D.A.T.); (O.I.G.)
- Life Improvement by Future Technology (LIFT) Center, 121205 Moscow, Russia
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
20
|
Muhammad N, Ajmera S, Lee JYK. Intraoperative visualization of cranial nerve schwannomas using second-window indocyanine green: A case series. Clin Neurol Neurosurg 2024; 240:108241. [PMID: 38522224 DOI: 10.1016/j.clineuro.2024.108241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/10/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Second Window Indocyanine Green (SWIG) is a novel intraoperative imaging technique that uses near-infrared (NIR) light for intra-operative tumor visualization using the well-known fluorophore indocyanine green (ICG). Because schwannomas often incorporate the nerve into the encapsulated tumor and impinge on surrounding neural structures, SWIG is a promising technique to improve tumor resection while sparing the nerve. OBJECTIVE To demonstrate the use of SWIG in resection of cranial nerve schwannomas. METHODS Three patients with cranial nerve schwannomas (i.e., trigeminal, vestibular, and vagus) underwent SWIG-guided resection. During surgery, NIR visualization was used intermittently used to detect fluorescence to guide resection. Signal-to-background ratio was then calculated to quantify fluorescence. RESULTS Patients were infused with ICG at a dose of 5.0 mg/kg 24 hours before surgery. Each patient achieved total or near-total resection and relief of symptoms with lack of recurrence at six-month follow-up. The average SBR calculated was 3.79, comparable to values for SWIG-guided resection of other brain and spine tumors. CONCLUSION This case series is the first published report of trigeminal and vagus nerve schwannoma resection using the SWIG technique and suggests that SWIG may be used to detect all schwannomas, alongside many other types of brain tumor. This paper also demonstrates the importance of preoperative ICG infusion timing and discusses the inverse pattern of NIR signal that may be observed when infusion occurs outside of the optimal timing. This provides direction for future studies investigating the administration of SWIG to resect cranial nerve schwannomas and other brain tumors.
Collapse
Affiliation(s)
- Najib Muhammad
- Department of Neurosurgery, Hospital of the University of Pennsylvania, 801 Spruce St, Philadelphia, PA 19107, USA
| | - Sonia Ajmera
- Department of Neurosurgery, Hospital of the University of Pennsylvania, 801 Spruce St, Philadelphia, PA 19107, USA
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, 801 Spruce St, Philadelphia, PA 19107, USA.
| |
Collapse
|
21
|
Chirizzi C, Pellegatta S, Gori A, Falco J, Rubiu E, Acerbi F, Bombelli FB. Next-generation agents for fluorescence-guided glioblastoma surgery. Bioeng Transl Med 2024; 9:e10608. [PMID: 38818124 PMCID: PMC11135154 DOI: 10.1002/btm2.10608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/09/2023] [Accepted: 09/05/2023] [Indexed: 06/01/2024] Open
Abstract
Glioblastoma is a fast-growing and aggressive form of brain cancer. Even with maximal treatment, patients show a low median survival and are often subjected to a high recurrence incidence. The currently available treatments require multimodal management, including maximal safe surgical resection, followed by radiation and chemotherapy. Because of the infiltrative glioblastoma nature, intraoperative differentiation of cancer tissue from normal brain parenchyma is very challenging, and this accounts for the low rate of complete tumor resection. For these reasons, clinicians have increasingly used various intraoperative adjuncts to improve surgical results, such as fluorescent agents. However, most of the existing fluorophores show several limitations such as poor selectivity, photostability, photosensitization and high costs. This could limit their application to successfully improve glioblastoma resection. In the present perspective, we highlight the possibility to develop next-generation fluorescent tools able to more selectively label cancer cells during surgical resection.
Collapse
Affiliation(s)
- Cristina Chirizzi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”Politecnico di MilanoMilanoItaly
| | - Serena Pellegatta
- Unit of Immunotherapy of Brain TumorsFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
- Unit of NeuroncologyFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Alessandro Gori
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC‐CNR)MilanItaly
| | - Jacopo Falco
- Neurosurgical Unit 2, Department of NeurosurgeryFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Emanuele Rubiu
- Neurosurgical Unit 2, Department of NeurosurgeryFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
| | - Francesco Acerbi
- Neurosurgical Unit 2, Department of NeurosurgeryFondazione IRCCS Istituto Neurologico Carlo BestaMilanItaly
- Experimental Microsurgical Laboratory, Department of NeurosurgeryFondazione IRCCS Istituto Neurologico Carlo BestaMilanoItaly
| | | |
Collapse
|
22
|
Gautheron A, Bernstock JD, Picart T, Guyotat J, Valdés PA, Montcel B. 5-ALA induced PpIX fluorescence spectroscopy in neurosurgery: a review. Front Neurosci 2024; 18:1310282. [PMID: 38348134 PMCID: PMC10859467 DOI: 10.3389/fnins.2024.1310282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/02/2024] [Indexed: 02/15/2024] Open
Abstract
The review begins with an overview of the fundamental principles/physics underlying light, fluorescence, and other light-matter interactions in biological tissues. It then focuses on 5-aminolevulinic acid (5-ALA)-induced protoporphyrin IX (PpIX) fluorescence spectroscopy methods used in neurosurgery (e.g., intensity, time-resolved) and in so doing, describe their specific features (e.g., hardware requirements, main processing methods) as well as their strengths and limitations. Finally, we review current clinical applications and future directions of 5-ALA-induced protoporphyrin IX (PpIX) fluorescence spectroscopy in neurosurgery.
Collapse
Affiliation(s)
- A. Gautheron
- Université Jean Monnet Saint-Etienne, CNRS, Institut d Optique Graduate School, Laboratoire Hubert Curien UMR 5516, Saint-Étienne, France
- Univ Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, CNRS, Inserm, CREATIS UMR 5220, U1294, Lyon, France
| | - J. D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - T. Picart
- Department of Neurosurgical Oncology and Vascular Neurosurgery, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon, Lyon, France
- Université Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - J. Guyotat
- Department of Neurosurgical Oncology and Vascular Neurosurgery, Pierre Wertheimer Neurological and Neurosurgical Hospital, Hospices Civils de Lyon, Lyon, France
| | - P. A. Valdés
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, United States
| | - B. Montcel
- Univ Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, CNRS, Inserm, CREATIS UMR 5220, U1294, Lyon, France
| |
Collapse
|
23
|
Chang CY, Chen CC. 5-aminolevulinic enhanced brain lesions mimic glioblastoma: A case report and literature review. Medicine (Baltimore) 2024; 103:e34518. [PMID: 38181251 PMCID: PMC10766299 DOI: 10.1097/md.0000000000034518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/06/2023] [Indexed: 01/07/2024] Open
Abstract
RATIONALE Glioblastoma multiforme (GBM) is a highly malignant primary brain tumor for which maximal tumor resection plays an important role in the treatment strategy. 5-aminolevulinic (5-ALA) is a powerful tool in fluorescence-guided surgery for GBM. However, 5-ALA- enhancing lesion can also be observed with different etiologies. PATIENTS CONCERNS Three cases of 5-ALA-enhancing lesions with etiologies different from glioma. DIAGNOSES The final diagnosis was abscess in 1 patient and diffuse large B-cell in the other 2 patients. INTERVENTIONS Three patients received 5-aminolevulinic acid-guided tumor resection under microscope with intraoperative neuromonitoring. OUTCOMES All of our patients showed improvement or stable neurological function outcomes. The final pathology revealed etiologies different from GBM. LESSONS The 5-aminolevulinic acid fluorescence-guided surgery has demonstrated its maximal extent of resection and safety profile in patients with high-grade glioma. Non-glioma etiologies may also mimic GBM in 5-ALA-guided surgeries. Therefore, patient history taking and consideration of brain images are necessary for the interpretation of 5-ALA-enhanced lesions.
Collapse
Affiliation(s)
- Chao-Yuan Chang
- Neurosurgical Department, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Chung Chen
- Neurosurgical Department, China Medical University Hospital, Taichung, Taiwan
- Department of Surgery, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
24
|
Al-Adli NN, Young JS, Scotford K, Sibih YE, Payne J, Berger MS. Advances in Intraoperative Glioma Tissue Sampling and Infiltration Assessment. Brain Sci 2023; 13:1637. [PMID: 38137085 PMCID: PMC10741454 DOI: 10.3390/brainsci13121637] [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: 10/12/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Gliomas are infiltrative brain tumors that often involve functional tissue. While maximal safe resection is critical for maximizing survival, this is challenged by the difficult intraoperative discrimination between tumor-infiltrated and normal structures. Surgical expertise is essential for identifying safe margins, and while the intraoperative pathological review of frozen tissue is possible, this is a time-consuming task. Advances in intraoperative stimulation mapping have aided surgeons in identifying functional structures and, as such, has become the gold standard for this purpose. However, intraoperative margin assessment lacks a similar consensus. Nonetheless, recent advances in intraoperative imaging techniques and tissue examination methods have demonstrated promise for the accurate and efficient assessment of tumor infiltration and margin delineation within the operating room, respectively. In this review, we describe these innovative technologies that neurosurgeons should be aware of.
Collapse
Affiliation(s)
- Nadeem N. Al-Adli
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94131, USA; (N.N.A.-A.); (J.S.Y.); (K.S.); (J.P.)
- School of Medicine, Texas Christian University, Fort Worth, TX 76109, USA
| | - Jacob S. Young
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94131, USA; (N.N.A.-A.); (J.S.Y.); (K.S.); (J.P.)
| | - Katie Scotford
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94131, USA; (N.N.A.-A.); (J.S.Y.); (K.S.); (J.P.)
| | - Youssef E. Sibih
- School of Medicine, University of California San Francisco, San Francisco, CA 94131, USA;
| | - Jessica Payne
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94131, USA; (N.N.A.-A.); (J.S.Y.); (K.S.); (J.P.)
| | - Mitchel S. Berger
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA 94131, USA; (N.N.A.-A.); (J.S.Y.); (K.S.); (J.P.)
| |
Collapse
|
25
|
Chen H, Mirg S, Gaddale P, Agrawal S, Li M, Nguyen V, Xu T, Li Q, Liu J, Tu W, Liu X, Drew PJ, Zhang N, Gluckman BJ, Kothapalli SR. Dissecting Multiparametric Cerebral Hemodynamics using Integrated Ultrafast Ultrasound and Multispectral Photoacoustic Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566048. [PMID: 37986863 PMCID: PMC10659547 DOI: 10.1101/2023.11.07.566048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Understanding brain-wide hemodynamic responses to different stimuli at high spatiotemporal resolutions can help study neuro-disorders and brain functions. However, the existing brain imaging technologies have limited resolution, sensitivity, imaging depth and provide information about only one or two hemodynamic parameters. To address this, we propose a multimodal functional ultrasound and photoacoustic (fUSPA) imaging platform, which integrates ultrafast ultrasound and multispectral photoacoustic imaging methods in a compact head-mountable device, to quantitatively map cerebral blood volume (CBV), cerebral blood flow (CBF), oxygen saturation (SO2) dynamics as well as contrast agent enhanced brain imaging with high spatiotemporal resolutions. After systematic characterization, the fUSPA system was applied to quantitatively study the changes in brain hemodynamics and vascular reactivity at single vessel resolution in response to hypercapnia stimulation. Our results show an overall increase in brain-wide CBV, CBF, and SO2, but regional differences in singular cortical veins and arteries and a reproducible anti-correlation pattern between venous and cortical hemodynamics, demonstrating the capabilities of the fUSPA system for providing multiparametric cerebrovascular information at high-resolution and sensitivity, that can bring insights into the complex mechanisms of neurodiseases.
Collapse
Affiliation(s)
- Haoyang Chen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Shubham Mirg
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Prameth Gaddale
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sumit Agrawal
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Menghan Li
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Van Nguyen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Tianbao Xu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Qiong Li
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jinyun Liu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Wenyu Tu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Xiao Liu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Institute for Computational and Data Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Patrick J. Drew
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Neurosurgery, The Pennsylvania State University, University Park, PA 16802, USA
| | - Nanyin Zhang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Bruce J. Gluckman
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Neurosurgery, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sri-Rajasekhar Kothapalli
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Penn State Cancer Institute, The Pennsylvania State University, Hershey, PA 17033, USA
- Graduate Program in Acoustics, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
26
|
Rainu SK, Ramachandran RG, Parameswaran S, Krishnakumar S, Singh N. Advancements in Intraoperative Near-Infrared Fluorescence Imaging for Accurate Tumor Resection: A Promising Technique for Improved Surgical Outcomes and Patient Survival. ACS Biomater Sci Eng 2023; 9:5504-5526. [PMID: 37661342 DOI: 10.1021/acsbiomaterials.3c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Clear surgical margins for solid tumor resection are essential for preventing cancer recurrence and improving overall patient survival. Complete resection of tumors is often limited by a surgeon's ability to accurately locate malignant tissues and differentiate them from healthy tissue. Therefore, techniques or imaging modalities are required that would ease the identification and resection of tumors by real-time intraoperative visualization of tumors. Although conventional imaging techniques such as positron emission tomography (PET), computed tomography (CT), magnetic resonance imaging (MRI), or radiography play an essential role in preoperative diagnostics, these cannot be utilized in intraoperative tumor detection due to their large size, high cost, long imaging time, and lack of cancer specificity. The inception of several imaging techniques has paved the way to intraoperative tumor margin detection with a high degree of sensitivity and specificity. Particularly, molecular imaging using near-infrared fluorescence (NIRF) based nanoprobes provides superior imaging quality due to high signal-to-noise ratio, deep penetration to tissues, and low autofluorescence, enabling accurate tumor resection and improved survival rates. In this review, we discuss the recent developments in imaging technologies, specifically focusing on NIRF nanoprobes that aid in highly specific intraoperative surgeries with real-time recognition of tumor margins.
Collapse
Affiliation(s)
- Simran Kaur Rainu
- Center for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Remya Girija Ramachandran
- L&T Ocular Pathology Department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai 600006, India
| | - Sowmya Parameswaran
- L&T Ocular Pathology Department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai 600006, India
| | - Subramanian Krishnakumar
- L&T Ocular Pathology Department, Vision Research Foundation, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Chennai 600006, India
| | - Neetu Singh
- Center for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
27
|
Borges HS, Gusmão LA, Tedesco AC. Multi-charged nanoemulsion for photodynamic treatment of glioblastoma cell line in 2D and 3D in vitro models. Photodiagnosis Photodyn Ther 2023; 43:103723. [PMID: 37487809 DOI: 10.1016/j.pdpdt.2023.103723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
Multi-charged nanoemulsions (NE) were designed to deliver Cannabidiol (CBD), Indocyanine green (ICG), and Protoporphyrin (PpIX) to treat glioblastoma (GBM) through Photodynamic Therapy (PDT). The phase-inversion temperature (PIT) method resulted in a highly stable NE that can be scaled easily, with a six-month shelf-life. We observed the quasi-spherical morphology of the nanoemulsions without any unencapsulated material and that 89% (± 5.5%) of the material was encapsulated. All physicochemical properties were within the expected range for a nanostructured drug delivery system, making these multi-charged nanoemulsions promising for further research and development. NE-PIC (NE-Protoporphyrin + Indocyanine + CBD) was easily internalized on GBM cells after three hours of incubation. Nanoemulsion (NE and NE-PIC) did not result in significant cytotoxicity, even for GBM or non-tumorigenic cell lines (NHF). Phototoxicity was significantly higher for the U87MG cell than the T98G cell when exposed to: visible (430 nm) and infrared (810 nm) laser light, with a difference of about 20%. From 50 mJ.cm-2, the viability of GBM cell lines decreases significantly, ranging from 65% to 85%. The NE-PIC was also effective for inhibiting cell proliferation into a 3D spheroidal GBM cell model, which is promising for mimicking the tumor cell environment. Irradiation at 810 nm was more effective in treating spheroid due to its deeper penetration in complex structures. NE-PIC has the potential as a drug delivery system for photoinactivation and photo diagnostic of GBM cell lines, taking advantage of the versatility of its active components.
Collapse
Affiliation(s)
- Hiago Salge Borges
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering ‒ Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901, Ribeirão Preto-SP, Brazil
| | - Luiza Araújo Gusmão
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering ‒ Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901, Ribeirão Preto-SP, Brazil
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering ‒ Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901, Ribeirão Preto-SP, Brazil.
| |
Collapse
|
28
|
Henderson F, Brem S, Hussain J, Buch L, Maloney E, Singhal S, Lee JYK. Second window indocyanine green localizes CNS lymphoma in real time in the operating room: report of two cases. Br J Neurosurg 2023; 37:619-623. [PMID: 32009484 PMCID: PMC10997215 DOI: 10.1080/02688697.2020.1716945] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 10/25/2022]
Abstract
Intraoperative distinction of lesional tissue versus normal brain parenchyma can be difficult in neurosurgical oncology procedures. We report the successful, real-time visualization of central nervous system (CNS) lymphoma using the 'Second Window Indocyanine Green' (SWIG) method for two patients who underwent craniotomy for pathology that was determined to be large B cell lymphoma. Indocyanine green (ICG), when administered intravenously the day prior to cranial surgery, is a re-purposed fluorophore that may afford safe, immediate visual confirmation of on-target tissue resection, thereby providing a valuable adjunct to intraoperative navigation and decreasing reliance on frozen pathology analysis. These first reported cases of SWIG for lymphoma in the CNS indicate that further study of fluorophores to improve biopsy targeting and yield is warranted.
Collapse
Affiliation(s)
- Fraser Henderson
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Steven Brem
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jasmin Hussain
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Love Buch
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Eileen Maloney
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
29
|
García-Montaño LA, Licón-Muñoz Y, Martinez FJ, Keddari YR, Ziemke MK, Chohan MO, Piccirillo SG. Dissecting Intra-tumor Heterogeneity in the Glioblastoma Microenvironment Using Fluorescence-Guided Multiple Sampling. Mol Cancer Res 2023; 21:755-767. [PMID: 37255362 PMCID: PMC10390891 DOI: 10.1158/1541-7786.mcr-23-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/25/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and recurrence, and improve the molecular classification of GBM.
Collapse
Affiliation(s)
- Leopoldo A. García-Montaño
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yamhilette Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Frank J. Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yasine R. Keddari
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of California, Merced, California
| | - Michael K. Ziemke
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Muhammad O. Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sara G.M. Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| |
Collapse
|
30
|
Belykh E, Bardonova L, Abramov I, Byvaltsev VA, Kerymbayev T, Yu K, Healey DR, Luna-Melendez E, Deneen B, Mehta S, Liu JK, Preul MC. 5-aminolevulinic acid, fluorescein sodium, and indocyanine green for glioma margin detection: analysis of operating wide-field and confocal microscopy in glioma models of various grades. Front Oncol 2023; 13:1156812. [PMID: 37287908 PMCID: PMC10242067 DOI: 10.3389/fonc.2023.1156812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/28/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction Surgical resection remains the first-line treatment for gliomas. Several fluorescent dyes are currently in use to augment intraoperative tumor visualization, but information on their comparative effectiveness is lacking. We performed systematic assessment of fluorescein sodium (FNa), 5-aminolevulinic acid (5-ALA)-induced protoporphyrin IX (PpIX), and indocyanine green (ICG) fluorescence in various glioma models using advanced fluorescence imaging techniques. Methods Four glioma models were used: GL261 (high-grade model), GB3 (low-grade model), and an in utero electroporation model with and without red fluorescence protein (IUE +RFP and IUE -RFP, respectively) (intermediate-to-low-grade model). Animals underwent 5-ALA, FNa, and ICG injections and craniectomy. Brain tissue samples underwent fluorescent imaging using a wide-field operative microscope and a benchtop confocal microscope and were submitted for histologic analysis. Results Our systematic analysis showed that wide-field imaging of highly malignant gliomas is equally efficient with 5-ALA, FNa, and ICG, although FNa is associated with more false-positive staining of the normal brain. In low-grade gliomas, wide-field imaging cannot detect ICG staining, can detect FNa in only 50% of specimens, and is not sensitive enough for PpIX detection. With confocal imaging of low-intermediate grade glioma models, PpIX outperformed FNa. Discussion Overall, compared to wide-field imaging, confocal microscopy significantly improved diagnostic accuracy and was better at detecting low concentrations of PpIX and FNa, resulting in improved tumor delineation. Neither PpIX, FNa, nor ICG delineated all tumor boundaries in studied tumor models, which emphasizes the need for novel visualization technologies and molecular probes to guide glioma resection. Simultaneous administration of 5-ALA and FNa with use of cellular-resolution imaging modalities may provide additional information for margin detection and may facilitate maximal glioma resection.
Collapse
Affiliation(s)
- Evgenii Belykh
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
- Department of Neurosurgery, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Liudmila Bardonova
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - Irakliy Abramov
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - Vadim A. Byvaltsev
- Department of Neurosurgery, Irkutsk State Medical University, Irkutsk, Russia
| | - Talgat Kerymbayev
- Department of Neurosurgery, JSC “National Scientific Center of Neurosurgery”, Nur-Sultan, Kazakhstan
| | - Kwanha Yu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States
| | - Debbie R. Healey
- Department of Research Imaging, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | | | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States
| | - Shwetal Mehta
- Ivy Brain Tumor Research Center, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| | - James K. Liu
- Department of Neurosurgery, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Mark C. Preul
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, United States
| |
Collapse
|
31
|
Wang N, Maswikiti EP, Wang B, Yu Y, Ma Y, Xiang L, Ma C, Ma Z, Gu B, Gao L, Chen H. Utility of near-infrared fluorescence imaging with indocyanine green in resection of oesophageal squamous cell carcinoma: A literature review and a case report. Photodiagnosis Photodyn Ther 2023; 42:103325. [PMID: 36746234 DOI: 10.1016/j.pdpdt.2023.103325] [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: 10/11/2022] [Revised: 01/26/2023] [Accepted: 02/03/2023] [Indexed: 02/06/2023]
Abstract
BACKGROUND Surgery remains the main primary treatment for non-advanced oesophageal cancer. Conventional thoracotomy and laparotomy can result in severe trauma, slow recovery, more complications, low quality of life, and reduced survival outcomes. Laparoscopic surgery has reduced the above-mentioned problems. However, some challenges remain associated with this approach, such as lymphadenectomy, anastomotic leakage, and inadequate surgical margins. Near-infrared fluorescence (NIRF) imaging using indocyanine green (ICG) in combination with laparoscopic surgery, provides real-time navigation throughout the entire surgical procedure. CASE PRESENTATION A middle-aged male patient presented to our health centre with progressive dysphagia for > 2 months. Endoscopy and biopsy revealed oesophageal squamous cell carcinoma 34 cm from the incisors (tumour node metastasis classification (TNM) T3N1M0 IIIB). ICG imaging fluorescence laparoscopic surgery was successfully performed to complete the oesophagectomy and oesophageal and tubular stomach anastomosis by accurately locating the lesion, retaining adequate upper and lower margins, visually dissecting the lymph nodes, and testing the anastomotic blood supply. The postoperative TNM stage was T2N0M0 ⅡA. The patient recovered quickly without complications. Postoperative chemotherapy was administered. After three years of follow-up, the patient had no recurrence or complications. CONCLUSIONS Fluorescence laparoscopy provides an excellent surgical treatment modality for patients with oesophageal cancer.
Collapse
Affiliation(s)
- Na Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Ewetse Paul Maswikiti
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Bofang Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Yang Yu
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Yanling Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Lin Xiang
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Chenhui Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Zhen Ma
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Baohong Gu
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| | - Lei Gao
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, 730030, China; Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Hao Chen
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China; Key Laboratory of System Tumors of Gansu Province, Lanzhou, Gansu, 730030, China.
| |
Collapse
|
32
|
Review of Intraoperative Adjuncts for Maximal Safe Resection of Gliomas and Its Impact on Outcomes. Cancers (Basel) 2022; 14:cancers14225705. [PMID: 36428797 PMCID: PMC9688206 DOI: 10.3390/cancers14225705] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Maximal safe resection is the mainstay of treatment in the neurosurgical management of gliomas, and preserving functional integrity is linked to favorable outcomes. How these modalities differ in their effectiveness on the extent of resection (EOR), survival, and complications remains unknown. A systematic literature search was performed with the following inclusion criteria: published between 2005 and 2022, involving brain glioma surgery, and including one or a combination of intraoperative modalities: intraoperative magnetic resonance imaging (iMRI), awake/general anesthesia craniotomy mapping (AC/GA), fluorescence-guided imaging, or combined modalities. Of 525 articles, 464 were excluded and 61 articles were included, involving 5221 glioma patients, 7(11.4%) articles used iMRI, 21(36.8%) used cortical mapping, 15(24.5%) used 5-aminolevulinic acid (5-ALA) or fluorescein sodium, and 18(29.5%) used combined modalities. The heterogeneity in reporting the amount of surgical resection prevented further analysis. Progression-free survival/overall survival (PFS/OS) were reported in 18/61(29.5%) articles, while complications and permanent disability were reported in 38/61(62.2%) articles. The reviewed studies demonstrate that intraoperative adjuncts such as iMRI, AC/GA mapping, fluorescence-guided imaging, and a combination of these modalities improve EOR. However, PFS/OS were underreported. Combining multiple intraoperative modalities seems to have the highest effect compared to each adjunct alone.
Collapse
|
33
|
Van Hese L, De Vleeschouwer S, Theys T, Rex S, Heeren RMA, Cuypers E. The diagnostic accuracy of intraoperative differentiation and delineation techniques in brain tumours. Discov Oncol 2022; 13:123. [PMID: 36355227 PMCID: PMC9649524 DOI: 10.1007/s12672-022-00585-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/22/2022] [Indexed: 11/11/2022] Open
Abstract
Brain tumour identification and delineation in a timeframe of seconds would significantly guide and support surgical decisions. Here, treatment is often complicated by the infiltration of gliomas in the surrounding brain parenchyma. Accurate delineation of the invasive margins is essential to increase the extent of resection and to avoid postoperative neurological deficits. Currently, histopathological annotation of brain biopsies and genetic phenotyping still define the first line treatment, where results become only available after surgery. Furthermore, adjuvant techniques to improve intraoperative visualisation of the tumour tissue have been developed and validated. In this review, we focused on the sensitivity and specificity of conventional techniques to characterise the tumour type and margin, specifically fluorescent-guided surgery, neuronavigation and intraoperative imaging as well as on more experimental techniques such as mass spectrometry-based diagnostics, Raman spectrometry and hyperspectral imaging. Based on our findings, all investigated methods had their advantages and limitations, guiding researchers towards the combined use of intraoperative imaging techniques. This can lead to an improved outcome in terms of extent of tumour resection and progression free survival while preserving neurological outcome of the patients.
Collapse
Affiliation(s)
- Laura Van Hese
- Division of Mass Spectrometry Imaging, Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- Department of Anaesthesiology, University Hospitals Leuven, 3000, Leuven, Belgium
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Steven De Vleeschouwer
- Neurosurgery Department, University Hospitals Leuven, 3000, Leuven, Belgium
- Laboratory for Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
| | - Tom Theys
- Neurosurgery Department, University Hospitals Leuven, 3000, Leuven, Belgium
- Laboratory for Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, Leuven Brain Institute (LBI), 3000, Leuven, Belgium
| | - Steffen Rex
- Department of Anaesthesiology, University Hospitals Leuven, 3000, Leuven, Belgium
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Ron M A Heeren
- Division of Mass Spectrometry Imaging, Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Eva Cuypers
- Division of Mass Spectrometry Imaging, Maastricht MultiModal Molecular Imaging (M4I) Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
34
|
Lew B, George M, Blair S, Zhu Z, Liang Z, Ludwig J, Kim CY, Kim KK, Gruev V, Choi H. Protease-activated indocyanine green nanoprobes for intraoperative NIR fluorescence imaging of primary tumors. NANOSCALE ADVANCES 2022; 4:4041-4050. [PMID: 36285222 PMCID: PMC9514568 DOI: 10.1039/d2na00276k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/25/2022] [Indexed: 05/17/2023]
Abstract
Tumor-targeted fluorescent probes in the near-infrared spectrum can provide invaluable information about the location and extent of primary and metastatic tumors during intraoperative procedures to ensure no residual tumors are left in the patient's body. Even though the first fluorescence-guided surgery was performed more than 50 years ago, it is still not accepted as a standard of care in part due to the lack of efficient and non-toxic targeted probes approved by regulatory agencies around the world. Herein, we report protease-activated cationic gelatin nanoparticles encapsulating indocyanine green (ICG) for the detection of primary breast tumors in murine models with high tumor-to-background ratios. Upon intravenous administration, these nanoprobes remain optically silent due to the energy resonance transfer among the bound ICG molecules. As the nanoprobes extravasate and are exposed to the acidic tumor microenvironment, their positive surface charges increase, facilitating cellular uptake. The internalized nanoprobes are activated upon proteolytic degradation of gelatin to allow high contrast between the tumor and normal tissue. Since both gelatin and ICG are FDA-approved for intravenous administration, this activatable nanoprobe can lead to quick clinical adoption and improve the treatment of patients undergoing image-guided cancer surgery.
Collapse
Affiliation(s)
- Benjamin Lew
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
| | - Mebin George
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
| | - Steven Blair
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
| | - Zhongmin Zhu
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
| | - Zuodong Liang
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
| | - Jamie Ludwig
- Division of Animal Resources, University of Illinois Urbana IL 61801 USA
| | - Celeste Y Kim
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
| | - Kyekyoon Kevin Kim
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
- Department of Bioengineering, University of Illinois Urbana IL 61801 USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana IL 61801 USA
| | - Viktor Gruev
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana IL 61801 USA
- Carle Illinois College of Medicine, University of Illinois Urbana IL 61801 USA
| | - Hyungsoo Choi
- Department of Electrical and Computer Engineering, University of Illinois Urbana IL 61801 USA
| |
Collapse
|
35
|
Rai H, Gupta S, Kumar S, Yang J, Singh SK, Ran C, Modi G. Near-Infrared Fluorescent Probes as Imaging and Theranostic Modalities for Amyloid-Beta and Tau Aggregates in Alzheimer's Disease. J Med Chem 2022; 65:8550-8595. [PMID: 35759679 DOI: 10.1021/acs.jmedchem.1c01619] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A person suspected of having Alzheimer's disease (AD) is clinically diagnosed for the presence of principal biomarkers, especially misfolded amyloid-beta (Aβ) and tau proteins in the brain regions. Existing radiotracer diagnostic tools, such as PET imaging, are expensive and have limited availability for primary patient screening and pre-clinical animal studies. To change the status quo, small-molecular near-infrared (NIR) probes have been rapidly developed, which may serve as an inexpensive, handy imaging tool to comprehend the dynamics of pathogenic progression in AD and assess therapeutic efficacy in vivo. This Perspective summarizes the biochemistry of Aβ and tau proteins and then focuses on structurally diverse NIR probes with coverages of their spectroscopic properties, binding affinity toward Aβ and tau species, and theranostic effectiveness. With the summarized information and perspective discussions, we hope that this paper may serve as a guiding tool for designing novel in vivo imaging fluoroprobes with theranostic capabilities in the future.
Collapse
Affiliation(s)
- Himanshu Rai
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| | - Sarika Gupta
- Molecular Science Laboratory, National Institute of Immunology, New Delhi-110067, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Jian Yang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Sushil K Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| | - Chongzhao Ran
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Gyan Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| |
Collapse
|
36
|
Vergeer RA, Theunissen REP, van Elk T, Schmidt I, Postma MR, Tamasi K, van Dijk JMC, Kuijlen JMA. Fluorescence-guided detection of pituitary neuroendocrine tumor (PitNET) tissue during endoscopic transsphenoidal surgery available agents, their potential, and technical aspects. Rev Endocr Metab Disord 2022; 23:647-657. [PMID: 35344185 PMCID: PMC9156450 DOI: 10.1007/s11154-022-09718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 10/29/2022]
Abstract
Differentiation of pituitary neuroendocrine tumor (PitNET) tissue from surrounding normal tissue during surgery is challenging. A number of fluorescent agents is available for visualization of tissue discrepancy, with the potential of improving total tumor resection. This review evaluates the availability, clinical and technical applicability of the various fluorescent agents within the field of pituitary surgery. According to PRISMA guidelines, a systematic review was performed to identify reports describing results of in vivo application of fluorescent agents. In this review, 15 publications were included. Sodium Fluorescein (FNa) was considered in two studies. The first study reported noticeable fluorescence in adenoma tissue, the second demonstrated the strongest fluorescence in non-functioning pituitary adenomas. 5-Aminolevulinic acid (5-ALA) was investigated in three studies. One study compared laser-based optical biopsy system (OBS) with photo-diagnostic filter (PD) and found that the OBS was able to detect all microadenomas, even when MRI was negative. The second study retrospectively analyzed twelve pituitary adenomas and found only one positive for fluorescence. The third investigated fifteen pituitary adenomas of which one displayed vague fluorescence. Indocyanine green (ICG) was researched in four studies with variable results. Second-Window ICG yielded no significant difference between functioning and non-functioning adenomas in one study, while a second study displayed 4 times higher fluorescence in tumor tissue than in normal tissue. In three studies, OTL38 showed potential in non-functioning pituitary adenomas. At present, evidence for fluorescent agents to benefit total resection of PitNETs is lacking. OTL38 can potentially serve as a selective fluorescent agent in non-functioning pituitary adenomas in the near future.
Collapse
Affiliation(s)
- Rob A Vergeer
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Robin E P Theunissen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Theodora van Elk
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mark R Postma
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Katalin Tamasi
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J Marc C van Dijk
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jos M A Kuijlen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
37
|
Netufo O, Connor K, Shiels LP, Sweeney KJ, Wu D, O’Shea DF, Byrne AT, Miller IS. Refining Glioblastoma Surgery through the Use of Intra-Operative Fluorescence Imaging Agents. Pharmaceuticals (Basel) 2022; 15:550. [PMID: 35631376 PMCID: PMC9143023 DOI: 10.3390/ph15050550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive adult brain tumour with a dismal 2-year survival rate of 26-33%. Maximal safe resection plays a crucial role in improving patient progression-free survival (PFS). Neurosurgeons have the significant challenge of delineating normal tissue from brain tumour to achieve the optimal extent of resection (EOR), with 5-Aminolevulinic Acid (5-ALA) the only clinically approved intra-operative fluorophore for GBM. This review aims to highlight the requirement for improved intra-operative imaging techniques, focusing on fluorescence-guided imaging (FGS) and the use of novel dyes with the potential to overcome the limitations of current FGS. The review was performed based on articles found in PubMed an.d Google Scholar, as well as articles identified in searched bibliographies between 2001 and 2022. Key words for searches included 'Glioblastoma' + 'Fluorophore'+ 'Novel' + 'Fluorescence Guided Surgery'. Current literature has favoured the approach of using targeted fluorophores to achieve specific accumulation in the tumour microenvironment, with biological conjugates leading the way. These conjugates target specific parts overexpressed in the tumour. The positive results in breast, ovarian and colorectal tissue are promising and may, therefore, be applied to intracranial neoplasms. Therefore, this design has the potential to produce favourable results in GBM by reducing the residual tumour, which translates to decreased tumour recurrence, morbidity and ultimately, mortality in GBM patients. Several preclinical studies have shown positive results with targeted dyes in distinguishing GBM cells from normal brain parenchyma, and targeted dyes in the Near-Infrared (NIR) emission range offer promising results, which may be valuable future alternatives.
Collapse
Affiliation(s)
- Oluwakanyinsolami Netufo
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 2, D02 YN77 Dublin, Ireland; (O.N.); (K.C.); (L.P.S.); (K.J.S.); (A.T.B.)
| | - Kate Connor
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 2, D02 YN77 Dublin, Ireland; (O.N.); (K.C.); (L.P.S.); (K.J.S.); (A.T.B.)
| | - Liam P. Shiels
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 2, D02 YN77 Dublin, Ireland; (O.N.); (K.C.); (L.P.S.); (K.J.S.); (A.T.B.)
| | - Kieron J. Sweeney
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 2, D02 YN77 Dublin, Ireland; (O.N.); (K.C.); (L.P.S.); (K.J.S.); (A.T.B.)
- National Centre for Neurosurgery, Beaumont Hospital, 9, D09 V2N0 Dublin, Ireland
| | - Dan Wu
- Department of Chemistry, Royal College of Surgeons in Ireland (RCSI), 2, D02 YN77 Dublin, Ireland; (D.W.); (D.F.O.)
| | - Donal F. O’Shea
- Department of Chemistry, Royal College of Surgeons in Ireland (RCSI), 2, D02 YN77 Dublin, Ireland; (D.W.); (D.F.O.)
| | - Annette T. Byrne
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 2, D02 YN77 Dublin, Ireland; (O.N.); (K.C.); (L.P.S.); (K.J.S.); (A.T.B.)
- National Pre-Clinical Imaging Centre (NPIC), 2, D02 YN77 Dublin, Ireland
| | - Ian S. Miller
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 2, D02 YN77 Dublin, Ireland; (O.N.); (K.C.); (L.P.S.); (K.J.S.); (A.T.B.)
- National Pre-Clinical Imaging Centre (NPIC), 2, D02 YN77 Dublin, Ireland
| |
Collapse
|
38
|
Lee S, Park CS, Yoon H. Nanoparticulate Photoluminescent Probes for Bioimaging: Small Molecules and Polymers. Int J Mol Sci 2022; 23:4949. [PMID: 35563340 PMCID: PMC9100005 DOI: 10.3390/ijms23094949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/22/2022] Open
Abstract
Recent interest in research on photoluminescent molecules due to their unique properties has played an important role in advancing the bioimaging field. In particular, small molecules and organic dots as probes have great potential for the achievement of bioimaging because of their desirable properties. In this review, we provide an introduction of probes consisting of fluorescent small molecules and polymers that emit light across the ultraviolet and near-infrared wavelength ranges, along with a brief summary of the most recent techniques for bioimaging. Since photoluminescence probes emitting light in different ranges have different goals and targets, their respective strategies also differ. Diverse and novel strategies using photoluminescence probes against targets have gradually been introduced in the related literature. Among recent papers (published within the last 5 years) on the topic, we here concentrate on the photophysical properties and strategies for the design of molecular probes, with key examples of in vivo photoluminescence research for practical applications. More in-depth studies on these probes will provide key insights into how to control the molecular structure and size/shape of organic probes for expanded bioimaging research and applications.
Collapse
Affiliation(s)
- Sanghyuck Lee
- Department of Polymer Engineering, Graduate School, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea;
| | - Chul Soon Park
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea;
| | - Hyeonseok Yoon
- Department of Polymer Engineering, Graduate School, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea;
- School of Polymer Science and Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| |
Collapse
|
39
|
Cho SS, Teng CW, De Ravin E, Singh YB, Lee JYK. Assessment and Comparison of Three Dimensional Exoscopes for Near-Infrared Fluorescence-Guided Surgery using Second-Window Indocyanine-Green. J Korean Neurosurg Soc 2022; 65:572-581. [PMID: 35418003 PMCID: PMC9271809 DOI: 10.3340/jkns.2021.0202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/23/2021] [Indexed: 11/27/2022] Open
Abstract
Objective Compared to microscopes, exoscopes have advantages in field-depth, ergonomics, and educational value. Exoscopes are especially well-poised for adaptation into fluorescence-guided surgery (FGS) due to their excitation source, light path, and image processing capabilities. We evaluated the feasibility of near-infrared FGS using a 3-dimensional (3D), 4 K exoscope with near-infrared fluorescence imaging capability. We then compared it to the most sensitive, commercially-available near-infrared exoscope system (3D and 960 p). In-vitro and intraoperative comparisons were performed.
Methods Serial dilutions of indocyanine-green (1–2000 μg/mL) were imaged with the 3D, 4 K Olympus Orbeye (system 1) and the 3D, 960 p VisionSense Iridium (system 2). Near-infrared sensitivity was calculated using signal-to-background ratios (SBRs). In addition, three patients with brain tumors were administered indocyanine-green and imaged with system 1, with two also imaged with system 2 for comparison.
Results Systems 1 and 2 detected near-infrared fluorescence from indocyanine green concentrations of >250 μg/L and >31.3 μg/L, respectively. Intraoperatively, system 1 visualized strong near-infrared fluorescence from two, strongly gadolinium-enhancing meningiomas (SBR=2.4, 1.7). The high-resolution, bright images were sufficient for the surgeon to appreciate the underlying anatomy in the near-infrared mode. However, system 1 was not able to visualize fluorescence from a weakly-enhancing intraparenchymal metastasis. In contrast, system 2 successfully visualized both the meningioma and the metastasis but lacked high resolution stereopsis.
Conclusion Three-dimensional exoscope systems provide an alternative visualization platform for both standard microsurgery and near-infrared fluorescent guided surgery. However, when tumor fluorescence is weak (i.e., low fluorophore uptake, deep tumors), highly sensitive near-infrared visualization systems may be required.
Collapse
Affiliation(s)
- Steve S Cho
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Clare W Teng
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Emma De Ravin
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Yash B Singh
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
40
|
Rynda AY, Olyushin VE, Rostovtsev DM, Zabrodskaya YM, Papayan GV. [Comparative analysis of 5-ALA and chlorin E6 fluorescence-guided navigation in malignant glioma surgery]. Khirurgiia (Mosk) 2022:5-14. [PMID: 35080821 DOI: 10.17116/hirurgia20220115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To analyze specificity and sensitivity of 5-ALA and chlorin E6 fluorescence-guided navigation in malignant glioma surgery. MATERIAL AND METHODS Fluorescence-guided navigation was analyzed in 50 patients (2 groups) with high-grade glioma. All patients were treated at the Polenov Russian Neurosurgery Institute. Chlorin E6 1 mg/kg intravenously (Photoditazin) was used as a fluorescence inducer in 25 patients (the 1st group), 5-ALA 20 mg/kg orally (Alasens) - in other 25 patients (the 2nd group). Each group included 10 patients with glioma grade III and 15 patients with glioma grade IV. Both groups were statistically representative (p>0.05). RESULTS In patients with glioma grade III, sensitivity of chlorin E6 fluorescence-guided navigation was 83.8%, 5-ALA fluorescence - 82.5%. Specificity was 66.7% and 64.1%, respectively. In patients with glioma grade IV, sensitivity was 87.7% for chlorin E6 and 88.3% for 5-ALA. Specificity was 85.2% and 88.1%, respectively. CONCLUSION Statistical analysis confirmed comparable high efficacy of both agents in surgery of malignant gliomas. Sensitivity and specificity of fluorescence-guided navigation with chlorin E6 and 5-ALA were similar (p>0.05).
Collapse
Affiliation(s)
- A Yu Rynda
- Polenov Russian Neurosurgery Institute of the Branch of the Almazov National Medical Research Center, St. Petersburg, Russia
| | - V E Olyushin
- Polenov Russian Neurosurgery Institute of the Branch of the Almazov National Medical Research Center, St. Petersburg, Russia
| | - D M Rostovtsev
- Polenov Russian Neurosurgery Institute of the Branch of the Almazov National Medical Research Center, St. Petersburg, Russia
| | - Yu M Zabrodskaya
- Polenov Russian Neurosurgery Institute of the Branch of the Almazov National Medical Research Center, St. Petersburg, Russia
| | - G V Papayan
- Polenov Russian Neurosurgery Institute of the Branch of the Almazov National Medical Research Center, St. Petersburg, Russia
| |
Collapse
|
41
|
Karsalia R, Cheng NH, Teng CW, Cho SS, Harmsen S, Lee JYK. Second window ICG predicts postoperative MRI gadolinium enhancement in high grade gliomas and brain metastases. NEUROSURGICAL FOCUS: VIDEO 2022; 6:V8. [PMID: 36284582 PMCID: PMC9555347 DOI: 10.3171/2021.10.focvid21204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/21/2021] [Indexed: 11/06/2022]
Abstract
A prospective trial evaluating the utility of second window indocyanine green (SWIG) in predicting postoperative MRI gadolinium enhancement was performed on high-grade gliomas (HGGs) and brain metastases. Compared to white light alone, SWIG demonstrated a higher sensitivity, negative predictive value, and accuracy in predicting residual neoplasm on MRI. The specificity of SWIG for predicting MRI enhancement was higher in HGGs than brain metastases. Clinically, near-infrared (NIR) imaging was better able to predict tumor recurrence than postoperative MRI. These results illustrate how SWIG is able to take advantage of gadolinium-like distribution properties to extravasate into the tumor microenvironment, enabling guidance in surgical resection.
The video can be found here: https://stream.cadmore.media/r10.3171/2021.10.FOCVID21204
Collapse
Affiliation(s)
- Ritesh Karsalia
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Nina H. Cheng
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia
- Drexel University College of Medicine, Philadelphia, Pennsylvania; and
| | - Clare W. Teng
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Steve S. Cho
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona
| | - Stefan Harmsen
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia
| | - John Y. K. Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
42
|
Farrakhova D, Romanishkin I, Maklygina Y, Bezdetnaya L, Loschenov V. Analysis of Fluorescence Decay Kinetics of Indocyanine Green Monomers and Aggregates in Brain Tumor Model In Vivo. NANOMATERIALS 2021; 11:nano11123185. [PMID: 34947534 PMCID: PMC8709123 DOI: 10.3390/nano11123185] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/26/2022]
Abstract
Spectroscopic approach with fluorescence time resolution allows one to determine the state of a brain tumor and its microenvironment via changes in the fluorescent dye's fluorescence lifetime. Indocyanine green (ICG) is an acknowledged infra-red fluorescent dye that self-assembles into stable aggregate forms (ICG NPs). ICG NPs aggregates have a tendency to accumulate in the tumor with a maximum accumulation at 24 h after systemic administration, enabling extended intraoperative diagnostic. Fluorescence lifetime analysis of ICG and ICG NPs demonstrates different values for ICG monomers and H-aggregates, indicating promising suitability for fluorescent diagnostics of brain tumors due to their affinity to tumor cells and stability in biological tissue.
Collapse
Affiliation(s)
- Dina Farrakhova
- Prokhorov General Physics Institute of the Russian Academy of Science, 119991 Moscow, Russia; (I.R.); (Y.M.); (V.L.)
- Correspondence: ; Tel.: +7-968-587-52-75
| | - Igor Romanishkin
- Prokhorov General Physics Institute of the Russian Academy of Science, 119991 Moscow, Russia; (I.R.); (Y.M.); (V.L.)
| | - Yuliya Maklygina
- Prokhorov General Physics Institute of the Russian Academy of Science, 119991 Moscow, Russia; (I.R.); (Y.M.); (V.L.)
| | - Lina Bezdetnaya
- Centre de Recherche en Automatique de Nancy, CNRS, Université de Lorraine, 54519 Vandoeuvre-lès-Nancy, France;
- Institut de Cancérologie de Lorraine, 54519 Vandoeuvre-lès-Nancy, France
| | - Victor Loschenov
- Prokhorov General Physics Institute of the Russian Academy of Science, 119991 Moscow, Russia; (I.R.); (Y.M.); (V.L.)
- Institute of Engineering Physics for Biomedicine, National Research Nuclear University MEPhI, 115409 Moscow, Russia
| |
Collapse
|
43
|
Schupper AJ, Price G, Hadjipanayis CG. Robotic-Assisted Digital Exoscope for Resection of Cerebral Metastases: A Case Series. Oper Neurosurg (Hagerstown) 2021; 21:436-444. [PMID: 34561708 DOI: 10.1093/ons/opab356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/04/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Surgical resection is the primary treatment for cerebral metastases with safe complete resection as the goal. The robotically assisted digital surgical exoscope is a novel system with advanced visualization methods with recent applications in neurosurgery. OBJECTIVE To evaluate the outcomes for patients with cerebral metastases undergoing resection with the surgical exoscope. METHODS Data were retrospectively collected from patients with cerebral metastases where resection was achieved with using the surgical exoscope from 2016 to 2020. Demographics, clinical, imaging, and operative and outcome findings were collected. The relationship between perioperative data and discharge disposition as well as progression-free survival (PFS) and 12 mo overall survival (OS) was assessed. RESULTS A total of 31 patients (19 males) with a median patient age 63 yr (range 38-80) were included. Average pre- and postoperative volumes were 18.1 cc and 0.75 cc, respectively. Mean depth of the resected lesions was 0.6 cm (range 0-3.6 cm). Complete resection was achieved in 64.5% of patients. The mean extent of resection was 96.7%, with 71.0% achieving PFS at 6 mo. Overall PFS rate was 58.1% and the OS rate at 12 mo was 83.9%. Neurological complications included motor (35.5%) and sensory (12.9%) deficits, with 12 patients reporting no postoperative symptoms. CONCLUSION The surgical exoscope can delineate tumor tissues with high resolution, as shown by a gross total resection achieved for the majority of cases in our series. Postoperative complications and patient outcomes were similar to those reported with use of the operative microscope. Use of the exoscope can provide optimal visualization and delineation of cerebral metastases.
Collapse
Affiliation(s)
- Alexander J Schupper
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York, USA
| | - Gabrielle Price
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York, USA
| | - Constantinos G Hadjipanayis
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York, USA
| |
Collapse
|
44
|
Tsen SWD, Springer LE, Sharmah Gautam K, Tang R, Liang K, Sudlow G, Kucharski A, Pham CTN, Achilefu S. Non-invasive monitoring of arthritis treatment response via targeting of tyrosine-phosphorylated annexin A2 in chondrocytes. Arthritis Res Ther 2021; 23:265. [PMID: 34696809 PMCID: PMC8543875 DOI: 10.1186/s13075-021-02643-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The development and optimization of therapies for rheumatoid arthritis (RA) is currently hindered by a lack of methods for early non-invasive monitoring of treatment response. Annexin A2, an inflammation-associated protein whose presence and phosphorylation levels are upregulated in RA, represents a potential molecular target for tracking RA treatment response. METHODS LS301, a near-infrared dye-peptide conjugate that selectively targets tyrosine 23-phosphorylated annexin A2 (pANXA2), was evaluated for its utility in monitoring disease progression, remission, and early response to drug treatment in mouse models of RA by fluorescence imaging. The intraarticular distribution and localization of LS301 relative to pANXA2 was determined by histological and immunohistochemical methods. RESULTS In mouse models of spontaneous and serum transfer-induced inflammatory arthritis, intravenously administered LS301 showed selective accumulation in regions of joint pathology including paws, ankles, and knees with positive correlation between fluorescent signal and disease severity by clinical scoring. Whole-body near-infrared imaging with LS301 allowed tracking of spontaneous disease remission and the therapeutic response after dexamethasone treatment. Histological analysis showed preferential accumulation of LS301 within the chondrocytes and articular cartilage in arthritic mice, and colocalization was observed between LS301 and pANXA2 in the joint tissue. CONCLUSIONS We demonstrate that fluorescence imaging with LS301 can be used to monitor the progression, remission, and early response to drug treatment in mouse models of RA. Given the ease of detecting LS301 with portable optical imaging devices, the agent may become a useful early treatment response reporter for arthritis diagnosis and drug evaluation.
Collapse
Affiliation(s)
- Shaw-Wei D Tsen
- Departments of Radiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Luke E Springer
- Division of Rheumatology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Krishna Sharmah Gautam
- Departments of Radiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Rui Tang
- Departments of Radiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Kexian Liang
- Departments of Radiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Gail Sudlow
- Departments of Radiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Amir Kucharski
- Departments of Radiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Christine T N Pham
- Division of Rheumatology, Washington University School of Medicine, St Louis, MO, 63110, USA.
| | - Samuel Achilefu
- Departments of Radiology, Washington University School of Medicine, St Louis, MO, 63110, USA.
- Departments of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, 63110, USA.
- Departments of Biomedical Engineering, Washington University School of Medicine, St Louis, MO, 63110, USA.
| |
Collapse
|
45
|
Abramov I, Dru AB, Belykh E, Park MT, Bardonova L, Preul MC. Redosing of Fluorescein Sodium Improves Image Interpretation During Intraoperative Ex Vivo Confocal Laser Endomicroscopy of Brain Tumors. Front Oncol 2021; 11:668661. [PMID: 34660258 PMCID: PMC8514872 DOI: 10.3389/fonc.2021.668661] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 09/13/2021] [Indexed: 12/15/2022] Open
Abstract
Background Fluorescein sodium (FNa) is a fluorescence agent used with a wide-field operating microscope for intraoperative guidance and with confocal laser endomicroscopy (CLE) to evaluate brain tissue. Susceptibility of FNa to degradation over time may affect CLE image quality during prolonged surgeries. This study describes improved characteristics of CLE images after intraoperative redosing with FNa. Methods A retrospective analysis was performed using CLE images obtained ex vivo from samples obtained during tumor resections with FNa-based fluorescence guidance with a wide-field operating microscope. The comparison groups included CLE images acquired after FNa redosing (redose imaging group), images from the same patients acquired after the initial FNa dose (initial-dose imaging group), and images from patients in whom redosing was not used (single-dose imaging group). A detailed assessment of image quality and interpretation regarding different FNa dosage and timing of imaging after FNa administration was conducted for all comparison groups. Results The brightest and most contrasting images were observed in the redose group compared to the initial-dose and single-dose groups (P<0.001). The decay of FNa signal negatively correlated with brightness (rho = -0.52, P<0.001) and contrast (rho = -0.57, P<0.001). Different doses of FNa did not significantly affect the brightness (P=0.15) or contrast (P=0.09) in CLE images. As the mean timing of imaging increased, the percentage of accurately diagnosed images decreased (P=0.03). Conclusions The decay of the FNa signal is directly associated with image brightness and contrast. The qualitative interpretation scores of images were highest for the FNa redose imaging group. Redosing with FNa to improve the utility of CLE imaging should be considered a safe and beneficial strategy during prolonged surgeries.
Collapse
Affiliation(s)
- Irakliy Abramov
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Alexander B Dru
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Evgenii Belykh
- Department of Neurosurgery, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Marian T Park
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Liudmila Bardonova
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Mark C Preul
- The Loyal and Edith Davis Neurosurgical Research Laboratory, Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| |
Collapse
|
46
|
Rynda AY, Zabrodskaya YM, Olyushin VE, Rostovtsev DM, Sokolova TV, Papayan GV. [Morphological evaluation of the effectiveness of fluorescence navigation with chlorin e6 in surgery for malignant gliomas]. Arkh Patol 2021; 83:13-20. [PMID: 34609799 DOI: 10.17116/patol20218305113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To evaluate the effectiveness of fluorescence navigation with chlorin e6 in surgery for malignant gliomas based on surgical material morphological and immunohistochemical data. MATERIAL AND METHODS The surgical material obtained from patients with high-grade (Grade III-IV) anaplastic glioma was examined. Along with histological examination, the proliferation marker Ki-67, the cell cycle transcription factor protein p53, and vascular endothelial growth factor (VEGF) were determined. RESULTS A significant direct correlation was found between the expression of Ki-67, p53, and VEGF and the fluorescence intensity of tumor tissues (p<0.05). CONCLUSION The technique of fluorescence navigation using chlorin e6 in comparative morphopathological analysis has confirmed its effectiveness in surgery for malignant gliomas.
Collapse
Affiliation(s)
- A Yu Rynda
- Prof. A.L. Polenov Russian Neurosurgical Institute - Branch V.A. Almazov National Medical Research Center of the Ministry of Health of Russia, Saint Petersburg, Russia
| | - Yu M Zabrodskaya
- Prof. A.L. Polenov Russian Neurosurgical Institute - Branch V.A. Almazov National Medical Research Center of the Ministry of Health of Russia, Saint Petersburg, Russia
| | - V E Olyushin
- Prof. A.L. Polenov Russian Neurosurgical Institute - Branch V.A. Almazov National Medical Research Center of the Ministry of Health of Russia, Saint Petersburg, Russia
| | - D M Rostovtsev
- Prof. A.L. Polenov Russian Neurosurgical Institute - Branch V.A. Almazov National Medical Research Center of the Ministry of Health of Russia, Saint Petersburg, Russia
| | - T V Sokolova
- Prof. A.L. Polenov Russian Neurosurgical Institute - Branch V.A. Almazov National Medical Research Center of the Ministry of Health of Russia, Saint Petersburg, Russia
| | - G V Papayan
- Prof. A.L. Polenov Russian Neurosurgical Institute - Branch V.A. Almazov National Medical Research Center of the Ministry of Health of Russia, Saint Petersburg, Russia
| |
Collapse
|
47
|
Yan F, Zhuang J, Yu Q, Dou Z, Jiang X, Tan S, Han Y, Wu X, Zang Y, Li C, Li J, Chen H, Hu L, Li X, Chen G. Strategy of De Novo Design toward First-In-Class Imaging Agents for Simultaneously Differentiating Glioma Boundary and Grades. ACS Sens 2021; 6:3330-3339. [PMID: 34448576 DOI: 10.1021/acssensors.1c01168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The extent of resection and tumor grade are two predominant prognostic factors for glioma. Fluorescent imaging is promising to facilitate accurate resection and simultaneous tumor grading. However, no probe fulfilling this task has been reported. Herein, we proposed a strategy of de novo design toward first-in-class fluorescent probes for simultaneously differentiating glioma boundary and grades. By bioinformatics analysis in combination with experimental validation, platelet-derived growth factor receptor β (PDGFRβ) was revealed as a promising biomarker for glioma imaging and grading. Then, fluorogenic probe PDGFP 1 was designed, guided by the structure-activity relationship study. Finally, the probe was demonstrated to stain glioma cells and tissues in the mice orthotopic glioma model with high selectivity over normal brain cells or tissues. Meanwhile, ex vivo experiments using patient-derived samples indicated that the fluorescence was significantly positively correlated with the tumor grades. This result highlighted the feasibility of the three-step de novo probe design strategy and suggested PDGFP 1 as a promising probe for simultaneously differentiating glioma boundary and grades, showing prospects of clinical translation.
Collapse
Affiliation(s)
- Feng Yan
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jianfeng Zhuang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qian Yu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Zhangqi Dou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefeng Jiang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shuyu Tan
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yifeng Han
- Department of Chemistry, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xinyan Wu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Cong Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huaijun Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Libin Hu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xin Li
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Gao Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
48
|
Shahein M, Prevedello DM, Beaumont TL, Ismail K, Nouby R, Palettas M, Prevedello LM, Otto BA, Carrau RL. The role of indocyanine green fluorescence in endoscopic endonasal skull base surgery and its imaging correlations. J Neurosurg 2021; 135:923-933. [PMID: 33186906 DOI: 10.3171/2020.6.jns192775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 06/18/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The use of endoscope-integrated indocyanine green (E-ICG) has recently been introduced in skull base surgery. The quantitative correlation between E-ICG and T1-weighted gadolinium-enhanced (T1WGd) images for skull base tumors has not been previously assessed, to the authors' knowledge. In this study, the authors investigated the indications for use and the limitations of E-ICG and sought to correlate the endoscopic fluorescence pattern with MRI contrast enhancement. METHODS Following IRB approval, 20 patients undergoing endoscopic endonasal skull base surgery between June 2017 and August 2018 were enrolled in the study. Tumor fluorescence was measured using a blue color value and blood fluorescence as a control. Signal intensities (SIs) of tumor T1WGd images were measured and the internal carotid artery (ICA) SI was used as a control. For pituitary adenoma, the pituitary gland fluorescence was also measured. The relationships between ICG fluorescence and MRI enhancement measurements were analyzed. RESULTS Data showed that in pituitary adenoma there was a strong correlation between the ratios of gland/blood fluorescence to gland/ICA SI (n = 8; r = 0.92; p = 0.001) and tumor/blood fluorescence to tumor/ICA SI (n = 9; r = 0.82; p = 0.006). In other pathologies there was a strong correlation between the ratios of tumor/blood fluorescence and tumor/ICA SI (n = 9; r = 0.74; p = 0.022). The ICG fluorescence allowed perfusion assessment of the pituitary gland as well as of the nasoseptal flaps. Visualization of the surrounding vasculature was also feasible. CONCLUSIONS Defining the indications and understanding the limitations are critical for the effective use of E-ICG. Tumor fluorescence seems to correlate with preoperative MRI contrast enhancement.
Collapse
Affiliation(s)
- Mostafa Shahein
- Departments of1Neurological Surgery
- 3Department of Neurosurgery, Aswan University Hospitals, Aswan University, Aswan, Egypt
| | | | | | - Khalid Ismail
- 3Department of Neurosurgery, Aswan University Hospitals, Aswan University, Aswan, Egypt
| | - Radwan Nouby
- 3Department of Neurosurgery, Aswan University Hospitals, Aswan University, Aswan, Egypt
| | | | - Luciano M Prevedello
- 5Radiology, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Bradley A Otto
- Departments of1Neurological Surgery
- 2Otolaryngology-Head and Neck Surgery
| | - Ricardo L Carrau
- Departments of1Neurological Surgery
- 2Otolaryngology-Head and Neck Surgery
| |
Collapse
|
49
|
Wu W, Klockow JL, Zhang M, Lafortune F, Chang E, Jin L, Wu Y, Daldrup-Link HE. Glioblastoma multiforme (GBM): An overview of current therapies and mechanisms of resistance. Pharmacol Res 2021; 171:105780. [PMID: 34302977 PMCID: PMC8384724 DOI: 10.1016/j.phrs.2021.105780] [Citation(s) in RCA: 358] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is a WHO grade IV glioma and the most common malignant, primary brain tumor with a 5-year survival of 7.2%. Its highly infiltrative nature, genetic heterogeneity, and protection by the blood brain barrier (BBB) have posed great treatment challenges. The standard treatment for GBMs is surgical resection followed by chemoradiotherapy. The robust DNA repair and self-renewing capabilities of glioblastoma cells and glioma initiating cells (GICs), respectively, promote resistance against all current treatment modalities. Thus, durable GBM management will require the invention of innovative treatment strategies. In this review, we will describe biological and molecular targets for GBM therapy, the current status of pharmacologic therapy, prominent mechanisms of resistance, and new treatment approaches. To date, medical imaging is primarily used to determine the location, size and macroscopic morphology of GBM before, during, and after therapy. In the future, molecular and cellular imaging approaches will more dynamically monitor the expression of molecular targets and/or immune responses in the tumor, thereby enabling more immediate adaptation of tumor-tailored, targeted therapies.
Collapse
Affiliation(s)
- Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Jessica L Klockow
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Michael Zhang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA; Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Famyrah Lafortune
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Edwin Chang
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Linchun Jin
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| | - Yang Wu
- Department of Neuropathology, Institute of Pathology, Technical University of Munich, Munich, Bayern 81675, Germany
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
50
|
Cho SS, Salinas R, De Ravin E, Teng CW, Li C, Abdullah KG, Buch L, Hussain J, Ahmed F, Dorsey J, Mohan S, Brem S, Singhal S, Lee JYK. Near-Infrared Imaging with Second-Window Indocyanine Green in Newly Diagnosed High-Grade Gliomas Predicts Gadolinium Enhancement on Postoperative Magnetic Resonance Imaging. Mol Imaging Biol 2021; 22:1427-1437. [PMID: 31712948 DOI: 10.1007/s11307-019-01455-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Intraoperative molecular imaging with tumor-targeting fluorophores offers real-time detection of neoplastic tissue. The second window indocyanine green (SWIG) technique relies on passive accumulation of indocyanine green (ICG), a near-infrared fluorophore, in neoplastic tissues. In this study, we explore the ability of SWIG to detect neoplastic tissue and to predict postoperative magnetic resonance imaging (MRI) findings intraoperatively. PROCEDURES Retrospective data were collected from 36 patients with primary high-grade gliomas (HGG) enrolled as part of a larger trial between October 2014 and October 2018. Patients received systemic ICG infusions at 2.5-5 mg/kg 24 h preoperatively. Near-infrared fluorescence was recorded throughout the case and from biopsy specimens. The presence/location of residual SWIG signal after resection was compared to the presence/location of residual gadolinium enhancement on postoperative MRI. The extent of resection was not changed based on near-infrared imaging. RESULTS All 36 lesions demonstrated strong near-infrared fluorescence (signal-to-background = 6.8 ± 2.2) and 100 % of tumors reaching the cortex were visualized before durotomy. In 78 biopsy specimens, near-infrared imaging demonstrated higher sensitivity and accuracy than white light for diagnosing neoplastic tissue intraoperatively. Furthermore, near-infrared imaging predicted gadolinium enhancement on postoperative MRI with 91 % accuracy, with visualization of residual enhancement as small as 0.3 cm3. Patients with no residual near-infrared signal after resection were significantly more likely to have complete resection on postoperative MRI (p value < 0.0001). CONCLUSIONS Intraoperative imaging with SWIG demonstrates highly sensitive detection of HGG tissue in real time. Furthermore, post-resection near-infrared imaging correlates with postoperative MRI. Overall, our findings suggest that SWIG can provide surgeons with MRI-like results in real time, potentially increasing resection rates.
Collapse
Affiliation(s)
- Steve S Cho
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Salinas
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Emma De Ravin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Clare W Teng
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Carrie Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Love Buch
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jasmin Hussain
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Fahad Ahmed
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jay Dorsey
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Brem
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|