1
|
Montaño AR, Masillati A, Szafran DA, Shams NA, Hubbell GE, Barth CW, Gibbs SL, Wang LG. Matrix-designed bright near-infrared fluorophores for precision peripheral nerve imaging. Biomaterials 2025; 319:123190. [PMID: 39987852 PMCID: PMC11932071 DOI: 10.1016/j.biomaterials.2025.123190] [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: 10/27/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
The FDA's recent approval of pafolacianine, the first molecular targeted contrast agent for fluorescence-guided surgery (FGS), signifies a remarkable milestone in precision medicine. This advance offers new hope for cancer patients by enabling guided removal of cancerous tissues, where completed surgical removal remains a consistent challenge without real-time intraoperative guidance. For optimal surgical outcomes, delicate nerve tissues must be preserved to maintain patient quality of life. Despite advances in the clinical translation pipeline, the development of clinically viable nerve-specific contrast agents for FGS remains a significant challenge. Herein, a medicinal chemistry-based matrix design strategy was applied to effectively generate a synthetic roadmap permitting management of nerve-specificity within the near-infrared (NIR) oxazine fluorophore family. Many of these newly developed fluorophores demonstrated robust nerve-specificity and superior safety profiles, while also offering spectral profiles that are compatible with the clinical surgical FGS infrastructure. Notably, improving observed brightness in vivo enabled exceptional visibility of buried nerve tissue, a priority during surgical procedures. Critically, the lead probe showed a large dosage safety window capable of generating substantial contrast at doses 100x lower than the maximum tolerated dose. Following clinical translation, such NIR nerve-specific fluorophores stand poised to significantly improve outcomes for surgical patients by improving identification and visualization of surface and buried nerve tissues in real time within the surgical arena.
Collapse
Affiliation(s)
- Antonio R Montaño
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Anas Masillati
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Dani A Szafran
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Nourhan A Shams
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Grace E Hubbell
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Connor W Barth
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA
| | - Summer L Gibbs
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA.
| | - Lei G Wang
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA.
| |
Collapse
|
2
|
van der Toom HFE, de Bruijn HS, Pauw RJ, Koljenović S, Koppes S, Robinson DJ, Keereweer S. Rigid Autofluorescence Imaging as a Tool for Identifying Cholesteatoma During Otologic Surgery: Initial Ex Vivo Findings. Otolaryngol Head Neck Surg 2025. [PMID: 40277164 DOI: 10.1002/ohn.1274] [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: 12/16/2024] [Revised: 03/14/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE This study aims to determine whether rigid autofluorescence imaging can differentiate cholesteatoma from surrounding tissues to reduce residual disease after cholesteatoma surgery. STUDY DESIGN Ex vivo proof-of-principle study. SETTING Erasmus University Medical Center, Rotterdam, the Netherlands. METHODS Autofluorescence signals of cholesteatoma, mucosa, and bone were measured using confocal microscopy to confirm distinguishable spectral differences. Subsequently, rigid autofluorescence imaging with specific filter settings (λe = 405 nm and λd > 570 nm) was applied to 14 resected surgical specimens to obtain signal intensity and tissue-specific fluorescence ratios. RESULTS Cholesteatoma matrix without keratin exhibited a significantly higher autofluorescence intensity compared to mucosa (P < .03), with a cholesteatoma-to-mucosa ratio of 2.15. Similarly, autofluorescence intensity was elevated in the matrix with keratin (ratio: 2.25, P = .03) and perimatrix with keratin (ratio: 2.29, P = .04) relative to mucosa. Perimatrix without keratin showed a nonsignificant trend (ratio: 1.85, P = .06). Although cholesteatoma and bone showed no significant difference, this is clinically less relevant as bone is easily identifiable during surgery. CONCLUSION Rigid autofluorescence imaging demonstrates significant potential for improving cholesteatoma surgery by reliably differentiating cholesteatoma matrix, both with and without accumulated keratin, from mucosa, showing 2.25- and 2.15-fold higher signal intensities, respectively. This technique could assist otologic surgeons in achieving more complete resections, thereby reducing residual disease rates while preserving surrounding structures. Future research should focus on optimizing the technology for in vivo application, particularly for detecting small cholesteatoma fragments, and further evaluate factors influencing specificity and sensitivity in clinical practice.
Collapse
Affiliation(s)
- Hylke F E van der Toom
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Henriette S de Bruijn
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Center for Optical Diagnostics and Therapy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert Jan Pauw
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Senada Koljenović
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Sjors Koppes
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dominic J Robinson
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Center for Optical Diagnostics and Therapy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Stijn Keereweer
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Zweedijk BE, Dalmeijer SWR, van Manen L, Galema HA, Lauwerends LJ, Abbasi H, Kremer B, Verhoef C, Robinson DJ, Koppes SA, Vahrmeijer AL, van der Vorst JR, Hilling DE, Keereweer S. Molecular-Targeted Fluorescence Lymph Node Imaging Could Play a Clinical Role in the Surgical Setting: A Systematic Review. Cancers (Basel) 2025; 17:1352. [PMID: 40282528 PMCID: PMC12025374 DOI: 10.3390/cancers17081352] [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: 03/17/2025] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
The lymphatic system plays a crucial role in the spread of solid tumors and is often the first site of metastasis, as cancer cells typically invade nearby lymph nodes (LN) before potentially spreading to other LNs through the lymphatic system and distant organs through the bloodstream [...].
Collapse
Affiliation(s)
- Bo E. Zweedijk
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, University Hospital Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands (H.A.); (D.J.R.)
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (C.V.)
| | - Sebastiaan W. R. Dalmeijer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, University Hospital Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands (H.A.); (D.J.R.)
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (C.V.)
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Labrinus van Manen
- Department of Otorhinolaryngology, Groene Hart Hospital, Bleulandweg 10, 2803 HG Gouda, The Netherlands
| | - Hidde A. Galema
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, University Hospital Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands (H.A.); (D.J.R.)
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (C.V.)
| | - Lorraine J. Lauwerends
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, University Hospital Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands (H.A.); (D.J.R.)
| | - Hamed Abbasi
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, University Hospital Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands (H.A.); (D.J.R.)
| | - Bernd Kremer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, University Hospital Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands (H.A.); (D.J.R.)
| | - Cornelis Verhoef
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (C.V.)
| | - Dominic J. Robinson
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, University Hospital Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands (H.A.); (D.J.R.)
| | - Sjors A. Koppes
- Department of Pathology, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Joost R. van der Vorst
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Denise E. Hilling
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (C.V.)
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Stijn Keereweer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, University Hospital Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands (H.A.); (D.J.R.)
| |
Collapse
|
4
|
Bateman LM, Streeter SS, Hebert KA, Parker DJ, Obando K, Moreno KSS, Zanazzi GJ, Barth CW, Wang LG, Gibbs SL, Henderson ER. Ex Vivo Human Tissue Functions as a Testing Platform for the Evaluation of a Nerve-Specific Fluorophore. Mol Imaging Biol 2025; 27:23-31. [PMID: 39658767 DOI: 10.1007/s11307-024-01968-0] [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: 07/29/2024] [Revised: 10/02/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
SIGNIFICANCE Selecting a nerve-specific lead fluorescent agent for translation in fluorescence-guided surgery is time-consuming and expensive. Preclinical fluorescent agent studies rely primarily on animal models, which are a critical component of preclinical testing, but these models may not predict fluorophore performance in human tissues. AIM The primary aim of this study was to evaluate and compare two preclinical models to test tissue-specific fluorophores based on discarded human tissues. The secondary aim was to use these models to determine the ability of a molecularly targeted fluorophore, LGW16-03, to label ex vivo human nerve tissues. APPROACH Patients undergoing standard-of-care transtibial or transfemoral amputation were consented and randomized to topical or systemic administration of LGW16-03 following amputation. After probe administration, nerves and background tissues were surgically resected and imaged to determine nerve fluorescence signal-to-background tissue ratio (SBR) and signal-to-noise ratio (SNR) metrics. Analysis of variance (ANOVA) determined statistical differences in metric means between administration cohorts and background tissue groups. Receiver operating characteristic (ROC) curve-derived statistics quantified the discriminatory performance of LGW16-03 fluorescence for labeling nerve tissues. RESULTS Tissue samples from 18 patients were analyzed. Mean nerve-to-adipose SBR was greater than nerve-to-muscle SBR (p = 0.001), but mean nerve-to-adipose SNR was not statistically different from mean nerve-to-muscle SNR (p = 0.069). Neither SBR nor SNR means were statistically different between fluorophore administration cohorts (p ≥ 0.448). When administration cohorts were combined, nerve-to-adipose SBR was greater than nerve-to-muscle SBR (mean ± standard deviation; 4.2 ± 2.9 vs. 1.8 ± 1.9; p < 0.001), but SNRs for nerve-to-adipose and nerve-to-muscle were not significantly different (5.1 ± 4.0 vs. 3.1 ± 3.4; p = 0.055). ROC curve-derived statistics to quantify LGW16-03 nerve labeling performance varied widely between patients, with sensitivities and specificities ranging from 0.2-99.9% and 0.4-100.0%. CONCLUSION Systemic and topical administration of LGW16-03 yielded similar fluorescence labeling of nerve tissues. Both administration approaches provided nerve-specific contrast similar to that observed in preclinical animal models. Fluorescence contrast was generally higher for nerve-to-adipose versus nerve-to-muscle. Ex vivo human tissue models provide safe evaluation of fluorophores in the preclinical phase and can aid in the selection of lead agents prior to first-in-human trials.
Collapse
Affiliation(s)
- Logan M Bateman
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, USA
| | - Samuel S Streeter
- Department of Orthopaedics, Dartmouth Health, Lebanon, NH, 03756, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, USA
| | - Kendra A Hebert
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Dylan J Parker
- Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, USA
| | - Kaye Obando
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
| | | | - George J Zanazzi
- Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, USA
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH, 03756, USA
| | - Connor W Barth
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Lei G Wang
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Summer L Gibbs
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Eric R Henderson
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA.
- Department of Orthopaedics, Dartmouth Health, Lebanon, NH, 03756, USA.
- Geisel School of Medicine, Dartmouth College, Hanover, NH, 03755, USA.
| |
Collapse
|
5
|
He P, Tang H, Zheng Y, Xu X, Peng X, Jiang T, Xiong Y, Zhang Y, Zhang Y, Liu G. Optical molecular imaging technology and its application in precise surgical navigation of liver cancer. Theranostics 2025; 15:1017-1034. [PMID: 39776802 PMCID: PMC11700863 DOI: 10.7150/thno.102671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/30/2024] [Indexed: 01/11/2025] Open
Abstract
Recent innovations in medical imaging technology have placed molecular imaging techniques at the forefront of diagnostic advancements. The current research trajectory in this field aims to integrate personalized molecular data of patients and diseases with traditional anatomical imaging data, enabling more precise, non-invasive, or minimally invasive diagnostic options for clinical medicine. This article provides an in-depth exploration of the basic principles and system components of optical molecular imaging technology. It also examines commonly used targeting mechanisms of optical probes, focusing especially on indocyanine green-the FDA-approved optical dye widely used in clinical settings-and its specific applications in diagnosing and treating liver cancer. Finally, this review highlights the advantages, limitations, and future challenges facing optical molecular imaging technology, offering a comprehensive overview of recent advances, clinical applications, and potential impacts on liver cancer treatment strategies.
Collapse
Affiliation(s)
- Pan He
- Department of Hepatobiliary and Pancreas Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
- Department of General Surgery, Institute of Hepatobiliary-Pancreatic-Intestinal Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Haitian Tang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Yating Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Xiao Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Xuqi Peng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Tao Jiang
- Department of General Surgery, Institute of Hepatobiliary-Pancreatic-Intestinal Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Yongfu Xiong
- Department of General Surgery, Institute of Hepatobiliary-Pancreatic-Intestinal Diseases, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Yang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| | - Yu Zhang
- Department of Hepatobiliary and Pancreas Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361002, China
| |
Collapse
|
6
|
Kriukova E, LaRochelle E, Pfefer TJ, Kanniyappan U, Gioux S, Pogue B, Ntziachristos V, Gorpas D. Impact of signal-to-noise ratio and contrast definition on the sensitivity assessment and benchmarking of fluorescence molecular imaging systems. JOURNAL OF BIOMEDICAL OPTICS 2025; 30:S13703. [PMID: 39034959 PMCID: PMC11256003 DOI: 10.1117/1.jbo.30.s1.s13703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024]
Abstract
Significance Standardization of fluorescence molecular imaging (FMI) is critical for ensuring quality control in guiding surgical procedures. To accurately evaluate system performance, two metrics, the signal-to-noise ratio (SNR) and contrast, are widely employed. However, there is currently no consensus on how these metrics can be computed. Aim We aim to examine the impact of SNR and contrast definitions on the performance assessment of FMI systems. Approach We quantified the SNR and contrast of six near-infrared FMI systems by imaging a multi-parametric phantom. Based on approaches commonly used in the literature, we quantified seven SNRs and four contrast values considering different background regions and/or formulas. Then, we calculated benchmarking (BM) scores and respective rank values for each system. Results We show that the performance assessment of an FMI system changes depending on the background locations and the applied quantification method. For a single system, the different metrics can vary up to ∼ 35 dB (SNR), ∼ 8.65 a . u . (contrast), and ∼ 0.67 a . u . (BM score). Conclusions The definition of precise guidelines for FMI performance assessment is imperative to ensure successful clinical translation of the technology. Such guidelines can also enable quality control for the already clinically approved indocyanine green-based fluorescence image-guided surgery.
Collapse
Affiliation(s)
- Elena Kriukova
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Technical University of Munich, School of Medicine and Health, Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Ethan LaRochelle
- QUEL Imaging, White River Junction, Vermont, United States
- Thayer School of Engineering at Dartmouth College, Hanover, New Hampshire, United States
| | - T. Joshua Pfefer
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland, United States
| | - Udayakumar Kanniyappan
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, Maryland, United States
| | - Sylvain Gioux
- Intuitive Surgical, Aubonne, Switzerland
- University of Strasbourg, ICube Laboratory, Strasbourg, France
| | - Brian Pogue
- University of Wisconsin Madison, Department of Medical Physics, Madison, Wisconsin, United States
| | - Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Technical University of Munich, School of Medicine and Health, Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), Munich, Germany
- Technical University of Munich, Munich Institute of Robotics and Machine Intelligence (MIRMI), Munich, Germany
| | - Dimitris Gorpas
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Technical University of Munich, School of Medicine and Health, Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), Munich, Germany
| |
Collapse
|
7
|
Barth C, Rizvi SZH, Masillati AM, Chackraborty S, Wang LG, Montaño AR, Szafran DA, Greer WS, van den Berg N, Sorger J, Rao DA, Alani AW, Gibbs SL. Nerve-Sparing Gynecologic Surgery Enabled by A Near-Infrared Nerve-Specific Fluorophore Using Existing Clinical Fluorescence Imaging Systems. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300011. [PMID: 37452434 PMCID: PMC11042870 DOI: 10.1002/smll.202300011] [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: 01/01/2023] [Revised: 05/09/2023] [Indexed: 07/18/2023]
Abstract
Patients undergoing gynecological procedures suffer from lasting side effects due to intraoperative nerve damage. Small, delicate nerves with complex and nonuniform branching patterns in the female pelvic neuroanatomy make nerve-sparing efforts during standard gynecological procedures such as hysterectomy, cystectomy, and colorectal cancer resection difficult, and thus many patients are left with incontinence and sexual dysfunction. Herein, a near-infrared (NIR) fluorescent nerve-specific contrast agent, LGW08-35, that is spectrally compatible with clinical fluorescence guided surgery (FGS) systems is formulated and characterized for rapid implementation for nerve-sparing gynecologic surgeries. The toxicology, pharmacokinetics (PK), and pharmacodynamics (PD) of micelle formulated LGW08-35 are examined, enabling the determination of the optimal imaging doses and time points, blood and tissue uptake parameters, and maximum tolerated dose (MTD). Application of the formulated fluorophore to imaging of female rat and swine pelvic neuroanatomy validates the continued clinical translation and use for real-time identification of important nerves such as the femoral, sciatic, lumbar, iliac, and hypogastric nerves. Further development of LGW08-35 for clinical use will unlock a valuable tool for surgeons in direct visualization of important nerves and contribute to the ongoing characterization of the female pelvic neuroanatomy to eliminate the debilitating side effects of nerve damage during gynecological procedures.
Collapse
Affiliation(s)
- Connor Barth
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
| | - Syed Zaki Husain Rizvi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, 97201
| | - Anas M. Masillati
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
| | - Samrat Chackraborty
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, 97201
| | - Lei G. Wang
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201
| | - Antonio R. Montaño
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
| | - Dani A. Szafran
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
| | - William S. Greer
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
| | | | | | - Deepa A. Rao
- School of Pharmacy, Pacific University, Hillsboro, OR 97123
| | - Adam W.G. Alani
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, 97201
| | - Summer L. Gibbs
- Biomedical Engineering Department, Oregon Health & Science University, Portland, OR 97201
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201
| |
Collapse
|
8
|
van Leeuwen FWB, Buckle T, van Oosterom MN, Rietbergen DDD. The Rise of Molecular Image-Guided Robotic Surgery. J Nucl Med 2024; 65:1505-1511. [PMID: 38991755 DOI: 10.2967/jnumed.124.267783] [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: 03/28/2024] [Accepted: 06/05/2024] [Indexed: 07/13/2024] Open
Abstract
Following early acceptance by urologists, the use of surgical robotic platforms is rapidly spreading to other surgical fields. This empowerment of surgical perception via robotic advances occurs in parallel to developments in intraoperative molecular imaging. Convergence of these efforts creates a logical incentive to advance the decades-old image-guided robotics paradigm. This yields new radioguided surgery strategies set to optimally exploit the symbiosis between the growing clinical translation of robotics and molecular imaging. These strategies intend to advance surgical precision by increasing dexterity and optimizing surgical decision-making. In this state-of-the-art review, topic-related developments in chemistry (tracer development) and engineering (medical device development) are discussed, and future scientific robotic growth markets for molecular imaging are presented.
Collapse
Affiliation(s)
- Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Daphne D D Rietbergen
- Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands; and
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
9
|
Collamati F, Morganti S, van Oosterom MN, Campana L, Ceci F, Luzzago S, Mancini-Terracciano C, Mirabelli R, Musi G, Nicolanti F, Orsi I, van Leeuwen FWB, Faccini R. First-in-human validation of a DROP-IN β-probe for robotic radioguided surgery: defining optimal signal-to-background discrimination algorithm. Eur J Nucl Med Mol Imaging 2024; 51:3098-3108. [PMID: 38376805 PMCID: PMC11300660 DOI: 10.1007/s00259-024-06653-6] [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: 11/22/2023] [Accepted: 02/07/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE In radioguided surgery (RGS), radiopharmaceuticals are used to generate preoperative roadmaps (e.g., PET/CT) and to facilitate intraoperative tracing of tracer avid lesions. Within RGS, there is a push toward the use of receptor-targeted radiopharmaceuticals, a trend that also has to align with the surgical move toward minimal invasive robotic surgery. Building on our initial ex vivo evaluation, this study investigates the clinical translation of a DROP-IN β probe in robotic PSMA-guided prostate cancer surgery. METHODS A clinical-grade DROP-IN β probe was developed to support the detection of PET radioisotopes (e.g., 68 Ga). The prototype was evaluated in 7 primary prostate cancer patients, having at least 1 lymph node metastases visible on PSMA-PET. Patients were scheduled for radical prostatectomy combined with extended pelvic lymph node dissection. At the beginning of surgery, patients were injected with 1.1 MBq/kg of [68Ga]Ga-PSMA. The β probe was used to trace PSMA-expressing lymph nodes in vivo. To support intraoperative decision-making, a statistical software algorithm was defined and optimized on this dataset to help the surgeon discriminate between probe signals coming from tumors and healthy tissue. RESULTS The DROP-IN β probe helped provide the surgeon with autonomous and highly maneuverable tracer detection. A total of 66 samples (i.e., lymph node specimens) were analyzed in vivo, of which 31 (47%) were found to be malignant. After optimization of the signal cutoff algorithm, we found a probe detection rate of 78% of the PSMA-PET-positive samples, a sensitivity of 76%, and a specificity of 93%, as compared to pathologic evaluation. CONCLUSION This study shows the first-in-human use of a DROP-IN β probe, supporting the integration of β radio guidance and robotic surgery. The achieved competitive sensitivity and specificity help open the world of robotic RGS to a whole new range of radiopharmaceuticals.
Collapse
Affiliation(s)
| | - Silvio Morganti
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lorenzo Campana
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Scienze di Base e Applicate per l'Ingegneria (SBAI), Sapienza University of Rome, Rome, Italy
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
| | - Stefano Luzzago
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Carlo Mancini-Terracciano
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Riccardo Mirabelli
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy.
- Department of Scienze di Base e Applicate per l'Ingegneria (SBAI), Sapienza University of Rome, Rome, Italy.
| | - Gennaro Musi
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Francesca Nicolanti
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Ilaria Orsi
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Riccardo Faccini
- National Institute of Nuclear Physics (INFN), Section of Rome, Rome, Italy
- Department of Physics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Berrens AC, Scheltema M, Maurer T, Hermann K, Hamdy FC, Knipper S, Dell'Oglio P, Mazzone E, de Barros HA, Sorger JM, van Oosterom MN, Stricker PD, van Leeuwen PJ, Rietbergen DDD, Valdes Olmos RA, Vidal-Sicart S, Carroll PR, Buckle T, van der Poel HG, van Leeuwen FWB. Delphi consensus project on prostate-specific membrane antigen (PSMA)-targeted surgery-outcomes from an international multidisciplinary panel. Eur J Nucl Med Mol Imaging 2024; 51:2893-2902. [PMID: 38012448 DOI: 10.1007/s00259-023-06524-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) is increasingly considered as a molecular target to achieve precision surgery for prostate cancer. A Delphi consensus was conducted to explore expert views in this emerging field and to identify knowledge and evidence gaps as well as unmet research needs that may help change practice and improve oncological outcomes for patients. METHODS One hundred and five statements (scored by a 9-point Likert scale) were distributed through SurveyMonkey®. Following evaluation, a consecutive second round was performed to evaluate consensus (16 statements; 89% response rate). Consensus was defined using the disagreement index, assessed by the research and development project/University of California, Los Angeles appropriateness method. RESULTS Eighty-six panel participants (72.1% clinician, 8.1% industry, 15.1% scientists, and 4.7% other) participated, most with a urological background (57.0%), followed by nuclear medicine (22.1%). Consensus was obtained on the following: (1) The diagnostic PSMA-ligand PET/CT should ideally be taken < 1 month before surgery, 1-3 months is acceptable; (2) a 16-20-h interval between injection of the tracer and surgery seems to be preferred; (3) PSMA targeting is most valuable for identification of nodal metastases; (4) gamma, fluorescence, and hybrid imaging are the preferred guidance technologies; and (5) randomized controlled clinical trials are required to define oncological value. Regarding surgical margin assessment, the view on the value of PSMA-targeted surgery was neutral or inconclusive. A high rate of "cannot answer" responses indicates further study is necessary to address knowledge gaps (e.g., Cerenkov or beta-emissions). CONCLUSIONS This Delphi consensus provides guidance for clinicians and researchers that implement or develop PSMA-targeted surgery technologies. Ultimately, however, the consensus should be backed by randomized clinical trial data before it may be implemented within the guidelines.
Collapse
Affiliation(s)
- Anne-Claire Berrens
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Matthijs Scheltema
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Center, Location VUmc, Amsterdam, The Netherlands
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ken Hermann
- Department of Nuclear Medicine, University of Duisburg-Essen, German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
- National Center for Tumor Diseases (NCT), NCT West, Heidelberg, Germany
| | - Freddie C Hamdy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Sophie Knipper
- Department of Urology, Vivantes Klinikum Am Urban, Berlin, Germany
| | - Paolo Dell'Oglio
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Urology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Elio Mazzone
- Unit of Urology/Division of Oncology, Gianfranco Soldera Prostate Cancer Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Hilda A de Barros
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | - Matthias N van Oosterom
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Philip D Stricker
- Department of Urology, St Vincents Hospital Sydney, Sydney, Australia
- St Vincents Prostate Cancer Research Center Sydney, Sydney, Australia
- Garvan Institute Sydney, Sydney, Australia
| | - Pim J van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Daphne D D Rietbergen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Renato A Valdes Olmos
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sergi Vidal-Sicart
- Department of Nuclear Medicine, Hospital Clínic Barcelona, Barcelona, Spain
| | - Peter R Carroll
- Department of Urology, University of California, San Francisco, CA, USA
| | - Tessa Buckle
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Center, Location VUmc, Amsterdam, The Netherlands
| | - Fijs W B van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
11
|
Ray GS, Streeter SS, Bateman LM, Elliott JT, Henderson ER. Real-time identification of life-threatening necrotizing soft-tissue infections using indocyanine green fluorescence imaging. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:066003. [PMID: 38745983 PMCID: PMC11092151 DOI: 10.1117/1.jbo.29.6.066003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
Significance Necrotizing soft-tissue infections (NSTIs) are life-threatening infections with a cumulative case fatality rate of 21%. The initial presentation of an NSTI is non-specific, frequently leading to misdiagnosis and delays in care. No current strategies yield an accurate, real-time diagnosis of an NSTI. Aim A first-in-kind, observational, clinical pilot study tested the hypothesis that measurable fluorescence signal voids occur in NSTI-affected tissues following intravenous administration and imaging of perfusion-based indocyanine green (ICG) fluorescence. This hypothesis is based on the established knowledge that NSTI is associated with local microvascular thrombosis. Approach Adult patients presenting to the Emergency Department of a tertiary care medical center at high risk for NSTI were prospectively enrolled and imaged with a commercial fluorescence imager. Single-frame fluorescence snapshot and first-pass perfusion kinetic parameters-ingress slope (IS), time-to-peak (TTP) intensity, and maximum fluorescence intensity (IMAX)-were quantified using a dynamic contrast-enhanced fluorescence imaging technique. Clinical variables (comorbidities, blood laboratory values), fluorescence parameters, and fluorescence signal-to-background ratios (SBRs) were compared to final infection diagnosis. Results Fourteen patients were enrolled and imaged (six NSTI, six cellulitis, one diabetes mellitus-associated gangrene, and one osteomyelitis). Clinical variables demonstrated no statistically significant differences between NSTI and non-NSTI patient groups (p -value ≥ 0.22 ). All NSTI cases exhibited prominent fluorescence signal voids in affected tissues, including tissue features not visible to the naked eye. All cellulitis cases exhibited a hyperemic response with increased fluorescence and no distinct signal voids. Median lesion-to-background tissue SBRs based on snapshot, IS, TTP, and IMAX parameter maps ranged from 3.2 to 9.1, 2.2 to 33.8, 1.0 to 7.5, and 1.5 to 12.7, respectively, for the NSTI patient group. All fluorescence parameters except TTP demonstrated statistically significant differences between NSTI and cellulitis patient groups (p -value < 0.05 ). Conclusions Real-time, accurate discrimination of NSTIs compared with non-necrotizing infections may be possible with perfusion-based ICG fluorescence imaging.
Collapse
Affiliation(s)
- Gabrielle S. Ray
- Dartmouth Health, Department of Orthopaedics, Lebanon, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
| | - Samuel S. Streeter
- Dartmouth Health, Department of Orthopaedics, Lebanon, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
| | - Logan M. Bateman
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Jonathan Thomas Elliott
- Dartmouth Health, Department of Orthopaedics, Lebanon, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Eric R. Henderson
- Dartmouth Health, Department of Orthopaedics, Lebanon, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - NEFARIOUS Study Group
- Dartmouth Health, Department of Orthopaedics, Lebanon, New Hampshire, United States
- Dartmouth College, Geisel School of Medicine, Hanover, New Hampshire, United States
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| |
Collapse
|
12
|
van Leeuwen FWB, Buckle T, Rietbergen DDD, van Oosterom MN. The realization of medical devices for precision surgery - development and implementation of ' stop-and-go' imaging technologies. Expert Rev Med Devices 2024; 21:349-358. [PMID: 38722051 DOI: 10.1080/17434440.2024.2341102] [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: 09/12/2023] [Accepted: 04/05/2024] [Indexed: 05/31/2024]
Abstract
INTRODUCTION Surgery and biomedical imaging encompass a big share of the medical-device market. The ever-mounting demand for precision surgery has driven the integration of these two into the field of image-guided surgery. A key-question herein is how imaging modalities can guide the surgical decision-making process. Through performance-based design, chemists, engineers, and doctors need to build a bridge between imaging technologies and surgical challenges. AREAS-COVERED This perspective article highlights the complementary nature between the technological design of an image-guidance modality and the type of procedure performed. The specific roles of the involved professionals, imaging technologies, and surgical indications are addressed. EXPERT-OPINION Molecular-image-guided surgery has the potential to advance pre-, intra- and post-operative tissue characterization. To achieve this, surgeons need the access to well-designed indication-specific chemical-agents and detection modalities. Hereby, some technologies stimulate exploration ('go'), while others stimulate caution ('stop'). However, failing to adequately address the indication-specific needs rises the risk of incorrect tool employment and sub-optimal surgical performance. Therefore, besides the availability of new technologies, market growth is highly dependent on the practical nature and impact on real-life clinical care. While urology currently takes the lead in the widespread implementation of image-guidance technologies, the topic is generic and its popularity spreads rapidly within surgical oncology.
Collapse
Affiliation(s)
- Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Daphne D D Rietbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Wang LG, Montaño AR, Masillati AM, Jones JA, Barth CW, Combs JR, Kumarapeli SU, Shams NA, van den Berg NS, Antaris AL, Galvis SN, McDowall I, Rizvi SZH, Alani AWG, Sorger JM, Gibbs SL. Nerve Visualization using Phenoxazine-Based Near-Infrared Fluorophores to Guide Prostatectomy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304724. [PMID: 37653576 DOI: 10.1002/adma.202304724] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/27/2023] [Indexed: 09/02/2023]
Abstract
Fluorescence-guided surgery (FGS) is poised to revolutionize surgical medicine through near-infrared (NIR) fluorophores for tissue- and disease-specific contrast. Clinical open and laparoscopic FGS vision systems operate nearly exclusively at NIR wavelengths. However, tissue-specific NIR contrast agents compatible with clinically available imaging systems are lacking, leaving nerve tissue identification during prostatectomy a persistent challenge. Here, it is shown that combining drug-like molecular design concepts and fluorophore chemistry enabled the production of a library of NIR phenoxazine-based fluorophores for intraoperative nerve-specific imaging. The lead candidate readily delineated prostatic nerves in the canine and iliac plexus in the swine using the clinical da Vinci Surgical System that has been popularized for minimally invasive prostatectomy procedures. These results demonstrate the feasibility of molecular engineering of NIR nerve-binding fluorophores for ready integration into the existing surgical workflow, paving the path for clinical translation to reduce morbidity from nerve injury for prostate cancer patients.
Collapse
Affiliation(s)
- Lei G Wang
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Antonio R Montaño
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Anas M Masillati
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Jocelyn A Jones
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Connor W Barth
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Jason R Combs
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
| | | | - Nourhan A Shams
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
| | | | | | - S N Galvis
- Intuitive Surgical, Sunnyvale, CA, 94086, USA
| | | | - Syed Zaki Husain Rizvi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, 97201, USA
| | - Adam W G Alani
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR, 97201, USA
| | | | - Summer L Gibbs
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR, 97201, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, 97201, USA
| |
Collapse
|
14
|
Pop CF, Veys I, Bormans A, Larsimont D, Liberale G. Fluorescence imaging for real-time detection of breast cancer tumors using IV injection of indocyanine green with non-conventional imaging: a systematic review of preclinical and clinical studies of perioperative imaging technologies. Breast Cancer Res Treat 2024; 204:429-442. [PMID: 38182824 PMCID: PMC10959791 DOI: 10.1007/s10549-023-07199-1] [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: 09/25/2023] [Accepted: 11/22/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND This review summarizes the available data on the effectiveness of indocyanine green fluorescence imaging (ICG-FI) for real-time detection of breast cancer (BC) tumors with perioperative imaging technologies. METHODS PubMed and Scopus databases were exhaustively searched for publications on the use of the real-time ICG-FI evaluation of BC tumors with non-conventional breast imaging technologies. RESULTS Twenty-three studies were included in this review. ICG-FI has been used for BC tumor identification in 12 orthotopic animal tumor experiences, 4 studies on animal assessment, and for 7 human clinical applications. The BC tumor-to-background ratio (TBR) was 1.1-8.5 in orthotopic tumor models and 1.4-3.9 in animal experiences. The detection of primary human BC tumors varied from 40% to 100%. The mean TBR reported for human BC varied from 2.1 to 3.7. In two studies evaluating BC surgical margins, good sensitivity (93.3% and 100%) and specificity (60% and 96%) have been reported, with a negative predictive value of ICG-FI to predict margin involvement intraoperatively of 100% in one study. CONCLUSIONS The use of ICG-FI as a guiding tool for the real-time identification of BC tumors and for the assessment of tumor boundaries is promising. There is great variability between the studies with regard to timing and dose. Further evidence is needed to assess whether ICG-guided BC surgery may be implemented as a standard of care.
Collapse
Affiliation(s)
- C Florin Pop
- Department of Surgical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Rue Meylemeersch 90, 1070, Brussels, Belgium.
| | - Isabelle Veys
- Department of Surgical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Rue Meylemeersch 90, 1070, Brussels, Belgium
| | - Anne Bormans
- Institutional Library, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Gabriel Liberale
- Department of Surgical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Rue Meylemeersch 90, 1070, Brussels, Belgium
| |
Collapse
|
15
|
Vreeburg MTA, Azargoshasb S, van Willigen D, Molenaar T, van Oosterom MN, Buckle T, Slof LJ, Klop M, Karakullukcu B, Donswijk M, van der Poel HG, van Leeuwen FWB, Brouwer OR, Rietbergen DDD. Comparison of two hybrid sentinel node tracers: indocyanine green (ICG)- 99mTc-nanocolloid vs. ICG- 99mTc-nanoscan from a nuclear medicine and surgical perspective. Eur J Nucl Med Mol Imaging 2023; 50:2282-2291. [PMID: 36929210 PMCID: PMC10250462 DOI: 10.1007/s00259-023-06157-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/15/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Lymph node (LN) metastasis is a relevant predictor for survival in patients with a.o. penile cancer (PeCa), malignant melanoma. The sentinel node (SN) procedure comprises targeted resection of the first tumour-draining SNs. Here, the hybrid tracer indocyanine green (ICG)-99mTc-nanocolloid has been used for several years to combine optical and nuclear detection. Recently, the resource of the nanocolloid precursor stopped production and the precursor was replaced by a different but chemically comparable colloid, nanoscan. Our aim was to study the performance of ICG-99mTc-nanoscan compared to ICG-99mTc-nanocolloid from a nuclear and surgical perspective. METHODS Twenty-four patients with either PeCa or head-and-neck (H&N) melanoma and scheduled for a SN procedure were included. The initial group (n = 11) received ICG-99mTc-nanocolloid until no longer available; the second group (n = 13) received ICG-99mTc-nanoscan. Tracer uptake was assessed on lymphoscintigraphy and single-photon emission (SPECT). Intraoperatively, SNs were identified using gamma tracing and fluorescence imaging. Ex vivo (back-table) measurements were conducted to quantify the fluorescence emissions. Chemical analysis was performed to compare the ICG assembly on both precursors. RESULTS The mean tracer uptake in the SNs was similar for ICG-99mTc-nanocolloid (2.2 ± 4.3%ID) and ICG-99mTc-nanoscan (1.8 ± 2.6%ID; p = 0.68). 3 SNs (interquartile range (IQR) 3-4) were detected on lymphoscintigraphy in PeCa patients receiving ICG-99mTc-nanoscan compared to 2 SNs (IQR 2-3) in PeCa patients receiving ICG-99mTc-nanocolloid (p = 0.045), no differences were observed in H&N patients. Back-table measurements of resected SNs revealed a lower total fluorescence intensity in the ICG-99mTc-nanoscan group (24*109 arbitrary units (A.U) IQR 1.6*109-14*109 in the ICG-99mTc-nanocolloid group versus 4.6*109 A.U. IQR 2.4*109-42*109 in the ICG-99mTc-nanoscan group, p = 0.0054). This was consistent with a larger degree of "stacked" ICG observed in the nanoscan formulation. No tracer-related adverse events were reported. CONCLUSIONS Based on this retrospective analysis, we can conclude that ICG-99mTc-nanoscan has similar capacity for SN identification as ICG-99mTc-nanocolloid and can safely be implemented in SN procedures.
Collapse
Affiliation(s)
- Manon T A Vreeburg
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Samaneh Azargoshasb
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Danny van Willigen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Tom Molenaar
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
- Radiochemistry Facility, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Matthias N van Oosterom
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Tessa Buckle
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Leon J Slof
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
- Instrumentele zaken ontwikkeling, facilitair bedrijf, Leiden University Medical Centre, Leiden, The Netherlands
| | - Martin Klop
- Department of Head and Neck Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Baris Karakullukcu
- Department of Head and Neck Surgery, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Maarten Donswijk
- Department of Nuclear Medicine, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Fijs W B van Leeuwen
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Oscar R Brouwer
- Department of Urology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands
| | - Daphne D D Rietbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Hospital, Leiden, The Netherlands.
- Department of Radiology, Section Nuclear Medicine, Leiden University Medical Hospital, Leiden, The Netherlands.
| |
Collapse
|
16
|
Wit EMK, KleinJan GH, Berrens AC, van Vliet R, van Leeuwen PJ, Buckle T, Donswijk ML, Bekers EM, van Leeuwen FWB, van der Poel HG. A hybrid radioactive and fluorescence approach is more than the sum of its parts; outcome of a phase II randomized sentinel node trial in prostate cancer patients. Eur J Nucl Med Mol Imaging 2023; 50:2861-2871. [PMID: 37036490 DOI: 10.1007/s00259-023-06191-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/05/2023] [Indexed: 04/11/2023]
Abstract
OBJECTIVE To determine the diagnostic accuracy of the hybrid tracer indocyanine green (ICG)-Technetium-99 m(99mTc)-nanocolloid compared to sequential tracers of 99mTc-nanocolloid and free-ICG in detecting tumor-positive lymph nodes (LN) during primary surgery in prostate cancer (PCa) patients. INTRODUCTION Image-guided surgery strategies can help visualize individual lymphatic drainage patterns and sentinel lymph nodes (SLNs) in PCa patients. For lymphatic mapping radioactive, fluorescent and hybrid tracers are being clinically exploited. In this prospective randomized phase II trial, we made a head-to-head comparison between ICG-99mTc-nanocolloid (hybrid group) and 99mTc-nanocolloid and subsequent free-ICG injection (sequential group). METHODS PCa patients with a >5% risk of lymphatic involvement according to the 2012 Briganti nomogram and planned for prostatectomy were included and randomized (1:1) between ultrasound-guided intraprostatic tracer administration of ICG-99mTc-nanocolloid (n = 69) or 99mTc-nanocolloid (n = 69) 5 h before surgery. Preoperative lymphoscintigraphy and SPECT/CT were performed to define the locations of the SLNs. Additionally, all participants in the sequential group received an injection of free-ICG at time of surgery. Subsequently, all (S)LNs were dissected using fluorescence guidance followed by an extended pelvic lymph node dissection (ePLND). The primary outcome was the total number of surgically removed (S)LNs and tumor-positive (S)LNs. RESULTS The total number of surgically removed (S)LN packages was 701 and 733 in the hybrid and sequential groups, respectively (p = 0.727). The total number of fluorescent LNs retrieved was 310 and 665 nodes in the hybrid and sequential groups, respectively (p < 0.001). However, no statistically significant difference was observed in the corresponding number of tumor-positive nodes among the groups (44 vs. 33; p = 0.470). Consequently, the rate of tumor-positive fluorescent LNs was higher in the hybrid group (7.4%) compared to the sequential group (2.6%; p = 0.002), indicating an enhanced positive predictive value for the hybrid approach. There was no difference in complications within 90 days after surgery (p = 0.78). CONCLUSIONS The hybrid tracer ICG-99mTc-nanocolloid improved the positive predictive value for tumor-bearing LNs while minimizing the number of fluorescent nodes compared to the sequential tracer approach. Consequently, the hybrid tracer ICG-99mTc-nanocolloid enables the most reliable and minimal invasive method for LN staging in PCa patients.
Collapse
Affiliation(s)
- Esther M K Wit
- Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Gijs H KleinJan
- Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Urology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne-Claire Berrens
- Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Roos van Vliet
- Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Pim J van Leeuwen
- Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Tessa Buckle
- Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maarten L Donswijk
- Department of Nuclear Medicine, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Elise M Bekers
- Department of Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Fijs W B van Leeuwen
- Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk G van der Poel
- Department of Urology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Berehova N, van Meerbeek MP, Azargoshasb S, van Willigen DM, Slof LJ, Navaei Lavasani S, van Oosterom MN, van Leeuwen FWB, Buckle T. A Truncated 14-Amino-Acid Myelin Protein-Zero-Targeting Peptide for Fluorescence-Guided Nerve-Preserving Surgery. Biomolecules 2023; 13:942. [PMID: 37371522 DOI: 10.3390/biom13060942] [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: 03/31/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The occurrence of accidental nerve damage during surgery and the increasing application of image guidance during head-and-neck surgery have highlighted the need for molecular targeted nerve-sparing interventions. The implementation of such interventions relies on the availability of nerve-specific tracers. In this paper, we describe the development of a truncated peptide that has an optimized affinity for protein zero (P0), the most abundant protein in myelin. METHODS AND MATERIALS Further C- and N-terminal truncation was performed on the lead peptide Cy5-P0101-125. The resulting nine Cy5-labelled peptides were characterized based on their photophysical properties, P0 affinity, and in vitro staining. These characterizations were combined with evaluation of the crystal structure of P0, which resulted in the selection of the optimized tracer Cy5-P0112-125. A near-infrared Cy7-functionalized derivative (Cy7-P0112-125) was used to perform an initial evaluation of fluorescence-guided surgery in a porcine model. RESULTS Methodological truncation of the 26-amino-acid lead compound Cy5-P0101-125 resulted in a size reduction of 53.8% for the optimized peptide Cy5-P0112-125. The peptide design and the 1.5-fold affinity gain obtained after truncation could be linked to interactions observed in the crystal structure of the extracellular portion of P0. The near-infrared analogue Cy7-P0112-125 supported nerve illumination during fluorescence-guided surgery in the head-and-neck region in a porcine model. CONCLUSIONS Methodological truncation yielded a second-generation P0-specific peptide. Initial surgical evaluation suggests that the peptide can support molecular targeted nerve imaging.
Collapse
Affiliation(s)
- Nataliia Berehova
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Maarten P van Meerbeek
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Samaneh Azargoshasb
- Design and Prototyping, Department of Medical Technology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Danny M van Willigen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Leon J Slof
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Design and Prototyping, Department of Medical Technology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Saaedeh Navaei Lavasani
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Head and Neck Surgery, Division of Surgical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Head and Neck Surgery, Division of Surgical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
18
|
Bortot B, Mangogna A, Di Lorenzo G, Stabile G, Ricci G, Biffi S. Image-guided cancer surgery: a narrative review on imaging modalities and emerging nanotechnology strategies. J Nanobiotechnology 2023; 21:155. [PMID: 37202750 DOI: 10.1186/s12951-023-01926-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023] Open
Abstract
Surgical resection is the cornerstone of solid tumour treatment. Current techniques for evaluating margin statuses, such as frozen section, imprint cytology, and intraoperative ultrasound, are helpful. However, an intraoperative assessment of tumour margins that is accurate and safe is clinically necessary. Positive surgical margins (PSM) have a well-documented negative effect on treatment outcomes and survival. As a result, surgical tumour imaging methods are now a practical method for reducing PSM rates and improving the efficiency of debulking surgery. Because of their unique characteristics, nanoparticles can function as contrast agents in image-guided surgery. While most image-guided surgical applications utilizing nanotechnology are now in the preclinical stage, some are beginning to reach the clinical phase. Here, we list the various imaging techniques used in image-guided surgery, such as optical imaging, ultrasound, computed tomography, magnetic resonance imaging, nuclear medicine imaging, and the most current developments in the potential of nanotechnology to detect surgical malignancies. In the coming years, we will see the evolution of nanoparticles tailored to specific tumour types and the introduction of surgical equipment to improve resection accuracy. Although the promise of nanotechnology for producing exogenous molecular contrast agents has been clearly demonstrated, much work remains to be done to put it into practice.
Collapse
Affiliation(s)
- Barbara Bortot
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Alessandro Mangogna
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giovanni Di Lorenzo
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Guglielmo Stabile
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giuseppe Ricci
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Stefania Biffi
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.
| |
Collapse
|
19
|
Zhang D, Teng KX, Zhao L, Niu LY, Yang QZ. Ultra-Small Nano-Assemblies as Tumor-Targeted and Renal Clearable Theranostic Agent for Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209789. [PMID: 36861334 DOI: 10.1002/adma.202209789] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/16/2023] [Indexed: 05/12/2023]
Abstract
It is a challenge to design photosensitizers to balance between the tumor-targeting enrichment for precise treatment and efficient clearance within a reasonable timescale for reducing side effects. Herein, an ultra-small nano-photosensitizer 1a with excellent tumor-specific accumulation and renal clearance is reported. It is formed from the self-assembly of compound 1 bearing three triethylene glycol (TEG) arms and two pyridinium groups in water. The positively charged surface with neutral TEG coating enables 1a to efficiently target the tumor, with the signal-to-background ratio reaching as high as 11.5 after tail intravenous injection. The ultra-small size of 1a with an average diameter of 5.6 nm allows its fast clearance through kidney. Self-assembly also endows 1a with an 18.2-fold enhancement of reactive oxygygen species generation rate compared to compound 1 in organic solution. Nano-PS 1a manifests an excellent photodynamic therapy efficacy on tumor-bearing mouse models. This work provides a promising design strategy of photosensitizers with renal clearable and tumor-targeting ability.
Collapse
Affiliation(s)
- Dongsheng Zhang
- Key Laboratory of Radiopharmaceuticals Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| | - Kun-Xu Teng
- Key Laboratory of Radiopharmaceuticals Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| | - Luyang Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Li-Ya Niu
- Key Laboratory of Radiopharmaceuticals Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| | - Qing-Zheng Yang
- Key Laboratory of Radiopharmaceuticals Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, P. R. China
| |
Collapse
|
20
|
Sikkenk DJ, Sterkenburg AJ, Schmidt I, Gorpas D, Nagengast WB, Consten ECJ. Detection of Tumour-Targeted IRDye800CW Tracer with Commercially Available Laparoscopic Surgical Systems. Diagnostics (Basel) 2023; 13:diagnostics13091591. [PMID: 37174982 PMCID: PMC10178288 DOI: 10.3390/diagnostics13091591] [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: 03/30/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
(1) Introduction: Near-infrared fluorescence (NIRF) combined with tumour-targeted tracers, such as bevacizumab-800CW, could aid surgical decision-making. This study explored the use of IRDye800CW, conjugated to bevacizumab, with four commercially available NIRF laparoscopes optimised for indocyanine green (ICG). (2) Methods: A (lymph node) phantom was made from a calibration device for NIRF and tissue-mimicking material. Serial dilutions of bevacizumab-800CW were made and ICG functioned as a reference. System settings, working distance, and thickness of tissue-mimicking material were varied to assess visibility of the fluorescence signal and tissue penetration. Tests were performed with four laparoscopes: VISERA ELITE II, Olympus; IMAGE1 S™ 4U Rubina, KARL STORZ; ENDOCAM Logic 4K platform, Richard Wolf; da Vinci Xi, Intuitive Surgical. (3) Results: The lowest visible bevacizumab-800CW concentration ranged between 13-850 nM (8-512 times diluted stock solution) for all laparoscopes, but the tracer was not visible through 0.8 cm of tissue in all systems. In contrast, ICG was still visible at a concentration of 0.4 nM (16,384 times diluted) and through 1.6-2.4 cm of tissue. Visibility and tissue penetration generally improved with a reduced working distance and manually adjusted system settings. (4) Conclusion: Depending on the application, bevacizumab-800CW might be sufficiently visible with current laparoscopes, but optimisation would widen applicability of tumour-targeted IRDye800CW tracers.
Collapse
Affiliation(s)
- Daan J Sikkenk
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Surgery, Meander Medical Centre, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| | - Andrea J Sterkenburg
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Dimitris Gorpas
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, Ismaninger Straße 22, D-81675 Munich, Germany
| | - Wouter B Nagengast
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Esther C J Consten
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Surgery, Meander Medical Centre, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| |
Collapse
|
21
|
Stibbe JA, de Barros HA, Linders DGJ, Bhairosingh SS, Bekers EM, van Leeuwen PJ, Low PS, Kularatne SA, Vahrmeijer AL, Burggraaf J, van der Poel HG. First-in-patient study of OTL78 for intraoperative fluorescence imaging of prostate-specific membrane antigen-positive prostate cancer: a single-arm, phase 2a, feasibility trial. Lancet Oncol 2023; 24:457-467. [PMID: 37062295 DOI: 10.1016/s1470-2045(23)00102-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND Targeted real-time imaging during robot-assisted radical prostatectomy provides information on the localisation and extent of prostate cancer. We assessed the safety and feasibility of the prostate-specific membrane antigen (PSMA)-targeted fluorescent tracer OTL78 in patients with prostate cancer. METHODS In this single-arm, phase 2a, feasibility trial with an adaptive design was carried out in The Netherlands Cancer Institute, Netherlands. Male patients aged 18 years or older, with PSMA PET-avid prostate cancer with an International Society of Urological Pathology (ISUP) grade group of 2 or more, who were scheduled to undergo robot-assisted radical prostatectomy with or without extended pelvic lymph node dissection were eligible. All patients had a robot-assisted radical prostatectomy using OTL78. Based on timing and dose, patients received a single intravenous infusion of OTL78 (0·06 mg/kg 1-2 h before surgery [dose cohort 1], 0·03 mg/kg 1-2 h before surgery [dose cohort 2], or 0·03 mg/kg 24 h before surgery [dose cohort 3]). The primary outcomes, assessed in all enrolled patients, were safety and pharmacokinetics of OTL78. This study is completed and is registered in the European Trial Database, 2019-002393-31, and the International Clinical Trials Registry Platform, NL8552, and is completed. FINDINGS Between June 29, 2020, and April 1, 2021, 19 patients were screened for eligibility, 18 of whom were enrolled. The median age was 69 years (IQR 64-70) and median prostate-specific antigen concentration was 15 ng/mL (IQR 9·3-22·0). In 16 (89%) of 18 patients, robot-assisted radical prostatectomy was accompanied by an extended pelvic lymph node dissection. Three serious adverse events occurred in one (6%) patient: an infected lymphocele, a urosepsis, and an intraperitoneal haemorrhage. These adverse events were considered unrelated to the administration of OTL78 or intraoperative fluorescence imaging. No patient died, required a dose reduction, or required discontinuation due to drug-related toxicity. The dose-normalised maximum serum concentration (Cmax/dose) in patients was 84·1 ng/mL/mg for the 0·03 mg/kg dose and 79·6 ng/mL/mg for the 0·06 mg/kg dose, the half-life was 5·1 h for the 0·03 mg/kg dose and 4·7 h for the 0·06 mg/kg dose, the volume of distribution was 22·9 L for the 0·03 mg/kg dose and 19·5 L for the 0·06 mg/kg dose, and the clearance was 3·1 L/h for the 0·03 mg/kg dose and 3·0 L/h for the 0·06 mg/kg dose. INTERPRETATION This first-in-patient study showed that OTL78 was well tolerated and had the potential to improve prostate cancer detection. Optimal dosing was 0·03 mg/kg, 24 h preoperatively. PSMA-directed fluorescence imaging allowed real-time identification of visually occult prostate cancer and might help to achieve complete oncological resections. FUNDING On Target Laboratories.
Collapse
Affiliation(s)
- Judith A Stibbe
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Hilda A de Barros
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Daan G J Linders
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Shadhvi S Bhairosingh
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Elise M Bekers
- Department of Pathology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Pim J van Leeuwen
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | | | | | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, Leiden University, Leiden, Netherlands; Centre for Human Drug Research, Leiden, Netherlands
| | - Henk G van der Poel
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands; Department of Urology, Amsterdam University Medical Center, Amsterdam, Netherlands.
| |
Collapse
|
22
|
Rijs Z, Belt E, Kalisvaart GM, Sier CFM, Kuppen PJK, Cleven AHG, Vahrmeijer AL, van de Sande MAJ, van Driel PBAA. Immunohistochemical Evaluation of Candidate Biomarkers for Fluorescence-Guided Surgery of Myxofibrosarcoma Using an Objective Scoring Method. Biomedicines 2023; 11:biomedicines11030982. [PMID: 36979961 PMCID: PMC10046284 DOI: 10.3390/biomedicines11030982] [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: 01/30/2023] [Revised: 02/22/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
INTRODUCTION Myxofibrosarcoma (MFS) is the most common soft-tissue sarcoma subtype in elderly patients. Local recurrence (LR) remains a major concern as the lack of intraoperative guidance and an infiltrative growth pattern with long, slender tails hamper surgeons' ability to achieve adequate resection margins for MFS. Fluorescence-guided surgery (FGS) could overcome this concern by delineating tumor tissue during surgery. One of the most important steps to successful FGS is to define a tumor-specific biomarker that is highly overexpressed in tumor tissue while low or absent in adjacent healthy tissue. The aim of this study is to evaluate the expression of eight previously selected promising biomarkers for FGS in MFS tissue samples with adjacent healthy tissue using immunohistochemistry (IHC). METHODS The following eight biomarkers were stained in seventeen paraffin-embedded MFS samples: tumor endothelial marker-1 (TEM-1, also known as endosialin/CD248), vascular endothelial growth factor receptor-1 (VEGFR-1, also known as Flt-1), vascular endothelial growth factor receptor-2 (VEGFR-2, also known as Flk1), vascular endothelial growth factor-A (VEGF-A), epidermal growth factor receptor (EGFR), insulin-like growth factor-1 receptor (IGF-1R), platelet derived growth factor receptor-α (PDGFR-α), and cluster of differentiation 40 (CD40, also known as TNFRSF5). A pathologist specializing in sarcoma annotated the margin between the tumor and adjacent healthy tissue in each MFS tissue sample. Subsequently, we used an objective IHC scoring method to assess and compare the difference in staining intensity between the tumor and adjacent healthy tissue, which is crucial for the use of FGS. RESULTS TEM-1, VEGF-A, and PDGFR-α stained all MFS tumors, while the other biomarkers did not show expression in all MFS tumors. Ultimately, TEM-1 was identified as the most suitable biomarker for FGS in MFS based on higher tumor-to-background (TBR) staining intensity compared to VEGF-A and PDGFR-α, regardless of preoperative therapy. CONCLUSION TEM-1-targeted FGS tracers should be further investigated to optimize MFS treatment.
Collapse
Affiliation(s)
- Zeger Rijs
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Esther Belt
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gijsbert M Kalisvaart
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Percuros BV, Zernikedreef 8, 2333 CL Leiden, The Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Arjen H G Cleven
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Pathology, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Michiel A J van de Sande
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | |
Collapse
|
23
|
Gibbs SL, Delikatny EJ. Editorial to the Special Issue Entitled "Optical Surgical Navigation". Mol Imaging Biol 2023; 25:1-2. [PMID: 36729349 DOI: 10.1007/s11307-023-01806-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Summer L Gibbs
- Biomedical Engineering Department, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97201, USA.
| | - Edward J Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
24
|
Berehova N, Buckle T, van Meerbeek MP, Bunschoten A, Velders AH, van Leeuwen FWB. Nerve Targeting via Myelin Protein Zero and the Impact of Dimerization on Binding Affinity. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27249015. [PMID: 36558148 PMCID: PMC9786614 DOI: 10.3390/molecules27249015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Surgically induced nerve damage is a common but debilitating side effect. By developing tracers that specifically target the most abundant protein in peripheral myelin, namely myelin protein zero (P0), we intend to support fluorescence-guided nerve-sparing surgery. To that end, we aimed to develop a dimeric tracer that shows a superior affinity for P0. METHODS Following truncation of homotypic P0 protein-based peptide sequences and fluorescence labeling, the lead compound Cy5-P0101-125 was selected. Using a bifunctional fluorescent dye, the dimeric Cy5-(P0101-125)2 was created. Assessment of the performance of the mono- and bi-labeled compounds was based on (photo)physical evaluation. This was followed by in vitro assessment in P0 expressing Schwannoma cell cultures by means of fluorescence confocal imaging (specificity, location of binding) and flow cytometry (binding affinity; KD). RESULTS Dimerization resulted in a 1.5-fold increase in affinity compared to the mono-labeled counterpart (70.3 +/- 10.0 nM vs. 104.9 +/- 16.7 nM; p = 0.003) which resulted in a 4-fold increase in staining efficiency in P0 expressing Schwannoma cells. Presence of two targeting vectors also improves a pharmacokinetics of labeled compounds by lowering serum binding and optical stability by preventing dye stacking. CONCLUSIONS Dimerization of the nerve-targeting peptide P0101-125 proves a valid strategy to improve P0 targeting.
Collapse
Affiliation(s)
- Nataliia Berehova
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Maarten P. van Meerbeek
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
| | - Anton Bunschoten
- Laboratory of BioNanoTechnology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Aldrik H. Velders
- Laboratory of BioNanoTechnology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
| | - Fijs W. B. van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC Leiden, The Netherlands
- Laboratory of BioNanoTechnology, Wageningen University & Research, Bornse Weilanden 9, 6708 WG Wageningen, The Netherlands
- Correspondence:
| |
Collapse
|
25
|
Boekestijn I, Azargoshasb S, van Oosterom MN, Slof LJ, Dibbets-Schneider P, Dankelman J, van Erkel AR, Rietbergen DDD, van Leeuwen FWB. Value-assessment of computer-assisted navigation strategies during percutaneous needle placement. Int J Comput Assist Radiol Surg 2022; 17:1775-1785. [PMID: 35934773 PMCID: PMC9468110 DOI: 10.1007/s11548-022-02719-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 07/04/2022] [Indexed: 11/05/2022]
Abstract
Abstract
Purpose
Navigational strategies create a scenario whereby percutaneous needle-based interventions of the liver can be guided using both pre-interventional 3D imaging datasets and dynamic interventional ultrasound (US). To score how such technologies impact the needle placement process, we performed kinematic analysis on different user groups.
Methods
Using a custom biopsy phantom, three consecutive exercises were performed by both novices and experts (n = 26). The exercise came in three options: (1) US-guidance, (2) US-guidance with pre-interventional image-registration (US + Reg) and (3) US-guidance with pre-interventional image-registration and needle-navigation (US + Reg + Nav). The traveled paths of the needle were digitized in 3D. Using custom software algorithms, kinematic metrics were extracted and related to dexterity, decision making indices to obtain overall performance scores (PS).
Results
Kinematic analysis helped quantifying the visual assessment of the needle trajectories. Compared to US-guidance, novices yielded most improvements using Reg (PSavg(US) = 0.43 vs. PSavg(US+Reg) = 0.57 vs. PSavg(US+Reg+Nav) = 0.51). Interestingly, the expert group yielded a reversed trend (PSavg(US) = 0.71 vs PSavg(US+Reg) = 0.58 vs PSavg(US+Reg+Nav) = 0.59).
Conclusion
Digitizing the movement trajectory allowed us to objectively assess the impact of needle-navigation strategies on percutaneous procedures. In particular, our findings suggest that these advanced technologies have a positive impact on the kinematics derived performance of novices.
Collapse
|