1
|
Won NJ, Bartling M, La Macchia J, Markevich S, Holtshousen S, Jagota A, Negus C, Najjar E, Wilson BC, Irish JC, Daly MJ. Deep learning-enabled fluorescence imaging for surgical guidance: in silico training for oral cancer depth quantification. JOURNAL OF BIOMEDICAL OPTICS 2025; 30:S13706. [PMID: 39295734 PMCID: PMC11408754 DOI: 10.1117/1.jbo.30.s1.s13706] [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: 04/30/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024]
Abstract
Significance Oral cancer surgery requires accurate margin delineation to balance complete resection with post-operative functionality. Current in vivo fluorescence imaging systems provide two-dimensional margin assessment yet fail to quantify tumor depth prior to resection. Harnessing structured light in combination with deep learning (DL) may provide near real-time three-dimensional margin detection. Aim A DL-enabled fluorescence spatial frequency domain imaging (SFDI) system trained with in silico tumor models was developed to quantify the depth of oral tumors. Approach A convolutional neural network was designed to produce tumor depth and concentration maps from SFDI images. Three in silico representations of oral cancer lesions were developed to train the DL architecture: cylinders, spherical harmonics, and composite spherical harmonics (CSHs). Each model was validated with in silico SFDI images of patient-derived tongue tumors, and the CSH model was further validated with optical phantoms. Results The performance of the CSH model was superior when presented with patient-derived tumors ( P -value < 0.05 ). The CSH model could predict depth and concentration within 0.4 mm and 0.4 μ g / mL , respectively, for in silico tumors with depths less than 10 mm. Conclusions A DL-enabled SFDI system trained with in silico CSH demonstrates promise in defining the deep margins of oral tumors.
Collapse
Affiliation(s)
- Natalie J Won
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Mandolin Bartling
- University of Toronto, Department of Otolaryngology-Head and Neck Surgery, Toronto, Ontario, Canada
| | - Josephine La Macchia
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Stefanie Markevich
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Scott Holtshousen
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Arjun Jagota
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Christina Negus
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Esmat Najjar
- University of Toronto, Department of Otolaryngology-Head and Neck Surgery, Toronto, Ontario, Canada
| | - Brian C Wilson
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- University of Toronto, Department of Medical Biophysics, Toronto, Ontario, Canada
| | - Jonathan C Irish
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- University of Toronto, Department of Otolaryngology-Head and Neck Surgery, Toronto, Ontario, Canada
| | - Michael J Daly
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Kim SY, Kim J, Kim H, Chang YT, Kwon HY, Lee JL, Yoon YS, Kim CW, Hong SM, Shin JH, Hong SW, Hwang SW, Ye BD, Byeon JS, Yang SK, Son BH, Myung SJ. Fluorescence-guided tumor visualization of colorectal cancer using tumor-initiating probe yellow in preclinical models. Sci Rep 2024; 14:26946. [PMID: 39505985 PMCID: PMC11542034 DOI: 10.1038/s41598-024-76312-1] [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: 04/25/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Fluorescence-guided surgery has emerged as an innovative technique with promising applications in the treatment of various tumors, including colon cancer. Tumor-initiating probe yellow (TiY) has been discovered for identifying tumorigenic cells by unbiased phenotypic screening with thousands of diversity-oriented fluorescence library (DOFL) compounds in a patient-derived lung cancer cell model. This study demonstrated the clinical feasibility of TiY for tumor-specific fluorescence imaging in the tissues of patients with colorectal cancer (CRC). To evaluate the efficacy of TiY in tumor imaging, surgical specimens were obtained, consisting of 36 tissues from 18 patients with CRC, for ex vivo molecular fluorescence imaging, histology, and immunohistochemistry. Orthotopic and chemically induced CRC mice models were administered TiY topically, and distinct tumor lesions were observed in 10 min by real-time fluorescence colonoscopy and ex vivo imaging. In a hepatic metastasis mouse model using splenic injection, TiY accumulation was detected in metastatic liver lesions through fluorescence imaging. Correlation analysis between TiY intensity and protein expression, assessed via immunohistochemistry and Western blotting, revealed a positive correlation between TiY and vimentin and Zeb1, which are known as epithelial-mesenchymal transition (EMT) markers of cancers. A comparative analysis of TiY with other FDA-approved fluorescence probes such as ICG revealed greater quantitative differences in TiY fluorescence intensity between tumor and normal tissues than those observed with ICG. Altogether, these results demonstrated that TiY has a strong potential for visualizing CRC by fluorescence imaging in various preclinical models, which can be further translated for clinical use such as fluorescence-guided surgery. Furthermore, our data indicate that TiY is preferentially uptaken by cells with EMT induction and progression, and overexpressing vimentin and Zeb1 in patients with CRC.
Collapse
Affiliation(s)
- Sun Young Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jinhyeon Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hajung Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Tae Chang
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Hwa-Young Kwon
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Jong Lyul Lee
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yong Sik Yoon
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chan Wook Kim
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Mo Hong
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Ho Shin
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung Wook Hong
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Wook Hwang
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byong Duk Ye
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Sik Byeon
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byung Ho Son
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Seung-Jae Myung
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Edis Biotech, Songpa-gu, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Jaiswal S, Cox KE, Amirfakhri S, Din Parast Saleh A, Kobayashi K, Lwin TM, Talib S, Aithal A, Mallya K, Jain M, Mohs AM, Hoffman RM, Batra SK, Bouvet M. Targeting Human Pancreatic Cancer with a Fluorophore-Conjugated Mucin 4 (MUC4) Antibody: Initial Characterization in Orthotopic Cell Line Mouse Models. J Clin Med 2024; 13:6211. [PMID: 39458160 PMCID: PMC11508345 DOI: 10.3390/jcm13206211] [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: 09/18/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Pancreatic cancer is the third leading cause of death related to cancer. The only possible cure presently is complete surgical resection; however, this is limited by difficulty in clearly defining tumor margins. Enhancement of the visualization of pancreatic ductal adenocarcinoma (PDAC) tumor margins using near-infrared dye-conjugated tumor-specific antibodies was pioneered by using anti-CEA, anti-CA19.9, and anti-MUC5AC in orthotopic mouse models of pancreatic cancer. Recently, an antibody to Mucin 4 (MUC4) conjugated to a fluorescent probe has shown promise in targeting colon tumors in orthotopic mouse models. Methods: In the present study, we targeted pancreatic cancer using an anti-MUC4 antibody conjugated to IRDye800 (anti-MUC4-IR800) in orthotopic mouse models. Two pancreatic cancer human cell lines were used, SW1990 and CD18/HPAF. Results: Anti-MUC4-IR800 targeted the two pancreatic cancer cell line tumors in orthotopic mouse models with high tumor-to-pancreas ratios and high tumor-to-liver ratios, with greater targeting seen in SW1990. Conclusions: The present results suggest anti-MUC4-IR800's potential to be used in fluorescence-guided surgical resection of pancreatic cancer.
Collapse
Affiliation(s)
- Sunidhi Jaiswal
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Kristin E. Cox
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Aylin Din Parast Saleh
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
| | - Keita Kobayashi
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
| | - Thinzar M. Lwin
- Department of Surgical Oncology, City of Hope, Duarte, CA 91010, USA;
| | - Sumbal Talib
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (S.T.); (A.M.M.)
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (A.A.); (K.M.); (M.J.); (S.K.B.)
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (A.A.); (K.M.); (M.J.); (S.K.B.)
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (A.A.); (K.M.); (M.J.); (S.K.B.)
| | - Aaron M. Mohs
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (S.T.); (A.M.M.)
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
- AntiCancer Inc., San Diego, CA 92111, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; (A.A.); (K.M.); (M.J.); (S.K.B.)
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA; (S.J.); (S.A.); (A.D.P.S.); (K.K.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
- UCSD Moores UCSD Cancer Center, 3855 Health Sciences Drive #0987, La Jolla, CA 92093-0987, USA
| |
Collapse
|
4
|
Shmuylovich L, O'Brien CM, Nwosu K, Achilefu S. Frugal engineering-inspired wearable augmented reality goggle system enables fluorescence-guided cancer surgery. Sci Rep 2024; 14:24402. [PMID: 39420102 PMCID: PMC11487067 DOI: 10.1038/s41598-024-75646-0] [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: 07/20/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Disparities in surgical outcomes often result from subjective decisions dictated by surgical training, experience, and available resources. To improve outcomes, surgeons have adopted advancements in robotics, endoscopy, and intra-operative imaging including fluorescence-guided surgery (FGS), which highlights tumors and anatomy in real-time. However, technical, economic, and logistic challenges hinder widespread adoption of FGS beyond high-resource centers. To overcome these impediments, we combined laser diodes, Raspberry Pi cameras and computers, off-the-shelf optical components, and 3D-printed parts to make a battery-powered, compact, dual white light and NIR imaging system that has comparable performance to existing bulkier, pricier, and wall-powered technologies. We combined these components with off-the-shelf augmented reality (AR) glasses to create a fully-wearable fluorescence imaging AR Raspberry Pi-based goggle system (FAR-Pi) and validated performance in a pre-clinical cancer surgery model. Novel device design ensures distance-independent coalignment between real and augmented views. As an open-source, affordable, and adaptable system, FAR-Pi is poised to democratize access to FGS and improve health outcomes worldwide.
Collapse
Affiliation(s)
- Leonid Shmuylovich
- Biophotonics Research Center, Department of Radiology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA.
- Division of Dermatology, Department of Medicine, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA.
| | - Christine M O'Brien
- Biophotonics Research Center, Department of Radiology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Karen Nwosu
- Biophotonics Research Center, Department of Radiology, Washington University in Saint Louis School of Medicine, Saint Louis, MO, USA
| | - Samuel Achilefu
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
5
|
Gupta S, Pal R, Schmidt EJ, Krishnamoorthy M, Leporati A, Kumar AT, Bogdanov A. Miniaturized Fab' imaging probe derived from a clinical antibody: Characterization and imaging in CRISPRi-attenuated mammary tumor models. iScience 2024; 27:110102. [PMID: 39184438 PMCID: PMC11342199 DOI: 10.1016/j.isci.2024.110102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/28/2024] [Accepted: 05/22/2024] [Indexed: 08/27/2024] Open
Abstract
Clinical imaging-assisted oncosurgical navigation requires cancer-specific miniaturized optical imaging probes. We report a near-infrared (NIR) Fab'-based epidermal growth factor receptor (EGFR)-specific probe carrying 3 NIR fluorophores (Fab'-800CW), which retained high-affinity binding to EGFR ectodomain (equilibrium KD E = 1 nM). Fab'-800CW showed a robust 4-times gain of fluorescence intensity (FI) and a 20% lifetime (FLT) increase under the conditions mimicking intracellular degradation. The probe was tested by using triple-negative breast cancer (TNBC) cell lines obtained by applying CRISPR interference (CRISPRi) effect of EGFR-targeting sgRNA and dCas9-KRAB chimera coexpression in MDA-MB-231 cells (WT cells). FI imaging in cell culture proved a 50% EGFR expression attenuation by CRISPRi. FI imaging in animals harboring attenuated or WT TNBC tumors with ex vivo corroboration identified differences between WT and CRISPRi tumors FI at 30 min post injection. Our results suggest the feasibility of EGFR expression imaging using a Fab'-based probe relevant for imaging-guided cancer surgery.
Collapse
Affiliation(s)
- Suresh Gupta
- Department of Radiology, UMASS Chan Medical School, Worcester, MA, USA
| | - Rahul Pal
- Mike Toth Head and Neck Cancer Research Center, Massachusetts Eye and Ear, Boston, MA, USA
- A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Eric J. Schmidt
- Department of Radiology, UMASS Chan Medical School, Worcester, MA, USA
| | - Murali Krishnamoorthy
- Mike Toth Head and Neck Cancer Research Center, Massachusetts Eye and Ear, Boston, MA, USA
- A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Anita Leporati
- Department of Radiology, UMASS Chan Medical School, Worcester, MA, USA
| | - Anand T.N. Kumar
- Mike Toth Head and Neck Cancer Research Center, Massachusetts Eye and Ear, Boston, MA, USA
- A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Alexei Bogdanov
- Department of Radiology, UMASS Chan Medical School, Worcester, MA, USA
- Cancer Center and Chemical Biology Interface Program, UMASS Chan Medical School, Worcester, MA, USA
| |
Collapse
|
6
|
Kennedy GT, Azari FS, Chang A, Bou-Samra P, Desphande C, Predina J, Delikatny EJ, Olson M, Rice DC, Singhal S. A Phase 2 Multicenter Clinical Trial of Intraoperative Molecular Imaging of Lung Cancer with a pH-Activatable Nanoprobe. Mol Imaging Biol 2024; 26:585-592. [PMID: 38992245 DOI: 10.1007/s11307-024-01933-x] [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: 05/14/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
PURPOSE Intraoperative molecular imaging (IMI) uses tumor-targeted optical contrast agents to improve identification and clearance of cancer. Recently, a probe has been developed that only fluoresces when activated in an acidic pH, which is common to many malignancies. We report the first multicenter Phase 2 trial of a pH-activatable nanoprobe (pegsitacianine, ONM-100) for IMI of lung cancer. METHODS Patients with suspected or biopsy-confirmed lung cancer scheduled for sublobar resection were administered a single intravenous infusion of pegsitacianine (1 mg/kg) one to three days prior to surgery. Intraoperatively, the patients underwent a white light thoracoscopic evaluation, and then were imaged with an NIR thoracoscope to detect tumor fluorescence. The primary study endpoint was the proportion of patients with a clinically significant event (CSE) which was defined as an intraoperative discovery during IMI that led to a change in the surgical procedure. Possible CSEs included (i) localizing the index lung nodule that could not be located by white light, (ii) identifying a synchronous malignant lesion, or (iii) recognizing a close surgical margin (< = 10 mm). Secondary endpoints were sensitivity, specificity, NPV, and PPV of pegsitacianine in detecting tumor-containing tissue. The safety evaluation was based on adverse event reporting, clinical laboratory parameters, and physical examinations. RESULTS Twenty patients were confirmed as eligible and administered pegsitacianine. Most of the patients were female (n = 12 [60%]), middle-aged (mean age 63.4 years), and former smokers (n = 13 [65%], 28.6 mean pack years). Mean lesion size was 1.9 cm, and most lesions (n = 17 [85%]) were malignant. The most common histologic subtype was adenocarcinoma (n = 9). By utilizing IMI with pegsitacianine, one patient had a CSE in the detection of a close margin and another had localization of a tumor not detectable by traditional surgical means. Six of 19 (31.6%) malignant lesions fluoresced with mean tumor-to-background ratio (TBR) of 3.00, as compared to TBR of 1.20 for benign lesions (n = 3). Sensitivity and specificity of pegsitacianine-based IMI for detecting malignant tissue was 31.6% and 33.3%, respectively. Positive predictive value (PPV) and negative predictive value (NPV) of pegsitacianine-based IMI was 75% and 7.1%, respectively. Pegsitacianine-based imaging was not effective in differentiating benign and malignant lymph nodes. From a safety perspective, no drug-related serious adverse events occurred. Four patients experienced mild pegsitacianine-related infusion reactions which required discontinuing the study drug with complete resolution of symptoms. CONCLUSIONS Pegsitacianine-based IMI, though well tolerated from a safety perspective, does not consistently label lung tumors during resection and does not provide significant clinical benefit over existing standards of surgical care. The biology of lung tumors may not be as acidic as other solid tumors in the body thereby not activating the probe as predicted.
Collapse
Affiliation(s)
- Gregory T Kennedy
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard 14th Floor South Pavilion, Philadelphia, PA, 19104, USA.
| | - Feredun S Azari
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard 14th Floor South Pavilion, Philadelphia, PA, 19104, USA
| | - Austin Chang
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard 14th Floor South Pavilion, Philadelphia, PA, 19104, USA
| | - Patrick Bou-Samra
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard 14th Floor South Pavilion, Philadelphia, PA, 19104, USA
| | - Charuhas Desphande
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Jarrod Predina
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard 14th Floor South Pavilion, Philadelphia, PA, 19104, USA
| | - Edward J Delikatny
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | - David C Rice
- Department of Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard 14th Floor South Pavilion, Philadelphia, PA, 19104, USA
| |
Collapse
|
7
|
Ram AS, Matuszewska K, McKenna C, Petrik J, Oblak ML. Validation of a semi-quantitative scoring system and workflow for analysis of fluorescence quantification in companion animals. Front Vet Sci 2024; 11:1392504. [PMID: 39144083 PMCID: PMC11322124 DOI: 10.3389/fvets.2024.1392504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024] Open
Abstract
Significance Many commercially available near-infrared (NIR) fluorescence imaging systems lack algorithms for real-time quantifiable fluorescence data. Creation of a workflow for clinical assessment and post hoc analysis may provide clinical researchers with a method for intraoperative fluorescence quantification to improve objective outcome measures. Aim Scoring systems and verified image analysis are employed to determine the amount and intensity of fluorescence within surgical specimens both intra and postoperatively. Approach Lymph nodes from canine cancer patients were obtained during lymph node extirpation following peritumoral injection of indocyanine green (ICG). First, a semi-quantitative assessment of surface fluorescence was evaluated. Images obtained with a NIR exoscope were analysed to determine fluorescence thresholds and measure fluorescence amount and intensity. Results Post hoc fluorescence quantification (threshold of Hue = 165-180, Intensity = 30-255) displayed strong agreement with semi-quantitative scoring (k = 0.9734, p < 0.0001). Fluorescence intensity with either threshold of 35-255 or 45-255 were significant predictors of fluorescence and had high sensitivity and specificity (p < 0.05). Fluorescence intensity and quantification had a strong association (p < 0.001). Conclusion The validation of the semi-quantitative scoring system by image analysis provides a method for objective in situ observation of tissue fluorescence. The utilization of thresholding for ICG fluorescence intensity allows post hoc quantification of fluorescence when not built into the imaging system.
Collapse
Affiliation(s)
- Ann S. Ram
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Kathy Matuszewska
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Charly McKenna
- Department of Clinical Studies, University of Guelph, Guelph, ON, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Michelle L. Oblak
- Department of Clinical Studies, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
8
|
Hart M, Isuri RK, Ramos D, Osharovich SA, Rodriguez AE, Harmsen S, Dudek GC, Huck JL, Holt DE, Popov AV, Singhal S, Delikatny EJ. Non-Small Cell Lung Cancer Imaging Using a Phospholipase A2 Activatable Fluorophore. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:490-500. [PMID: 39056064 PMCID: PMC11267604 DOI: 10.1021/cbmi.4c00026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 07/28/2024]
Abstract
Lung cancer, the most common cause of cancer-related death in the United States, requires advanced intraoperative detection methods to improve evaluation of surgical margins. In this study we employed DDAO-arachidonate (DDAO-A), a phospholipase A2 (PLA2) activatable fluorophore, designed for the specific optical identification of lung cancers in real-time during surgery. The in vitro fluorescence activation of DDAO-A by porcine sPLA2 was tested in various liposomal formulations, with 100 nm extruded EggPC showing the best overall characteristics. Extruded EggPC liposomes containing DDAO-A were tested for their stability under various storage conditions, demonstrating excellent stability for up to 4 weeks when stored at -20 °C or below. Cell studies using KLN 205 and LLC1 lung cancer cell lines showed DDAO-A activation was proportional to cell number. DDAO-A showed preferential activation by human recombinant cPLA2, an isoform highly specific to arachidonic acid-containing lipids, when compared to a control probe, DDAO palmitate (DDAO-P). In vivo studies with DBA/2 mice bearing KLN 205 lung tumors recapitulated these results, with preferential activation of DDAO-A relative to DDAO-P following intratumoral injection. Topical application of DDAO-A-containing liposomes to human (n = 10) and canine (n = 3) lung cancers ex vivo demonstrated the preferential activation of DDAO-A in tumor tissue relative to adjacent normal lung tissue, with fluorescent tumor-to-normal ratios (TNR) of up to 5.2:1. The combined results highlight DDAO-A as a promising candidate for clinical applications, showcasing its potential utility in intraoperative and back-table imaging and topical administration during lung cancer surgeries. By addressing the challenge of residual microscopic disease at resection margins and offering stability in liposomal formulations, DDAO-A emerges as a potentially valuable tool for advancing precision lung cancer surgery and improving curative resection rates.
Collapse
Affiliation(s)
- Michael
C. Hart
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Ritesh K. Isuri
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department
of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - Drew Ramos
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sofya A. Osharovich
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Andrea E. Rodriguez
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Stefan Harmsen
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Grace C. Dudek
- Department
of Biology, University of Pennsylvania, 102 Leidy Laboratories 433 S University
Ave, Philadelphia, Pennsylvania 19104, United States
| | - Jennifer L. Huck
- Department
of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - David E. Holt
- Department
of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Anatoliy V. Popov
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sunil Singhal
- Department
of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Edward J. Delikatny
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
9
|
Jeremiasse B, van Ineveld RL, Bok V, Kleinnijenhuis M, de Blank S, Alieva M, Johnson HR, van Vliet EJ, Zeeman AL, Wellens LM, Llibre-Palomar G, Barrera Román M, Di Maggio A, Dekkers JF, Oliveira S, Vahrmeijer AL, Molenaar JJ, Wijnen MH, van der Steeg AF, Wehrens EJ, Rios AC. A multispectral 3D live organoid imaging platform to screen probes for fluorescence guided surgery. EMBO Mol Med 2024; 16:1495-1514. [PMID: 38831131 PMCID: PMC11251264 DOI: 10.1038/s44321-024-00084-4] [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: 10/20/2023] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
Achieving complete tumor resection is challenging and can be improved by real-time fluorescence-guided surgery with molecular-targeted probes. However, pre-clinical identification and validation of probes presents a lengthy process that is traditionally performed in animal models and further hampered by inter- and intra-tumoral heterogeneity in target expression. To screen multiple probes at patient scale, we developed a multispectral real-time 3D imaging platform that implements organoid technology to effectively model patient tumor heterogeneity and, importantly, healthy human tissue binding.
Collapse
Affiliation(s)
- Bernadette Jeremiasse
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Ravian L van Ineveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Veerle Bok
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Michiel Kleinnijenhuis
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Sam de Blank
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Maria Alieva
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Instituto de Investigaciones Biomedicas Sols-Morreale (IIBM), CSIC-UAM, Madrid, Spain
| | - Hannah R Johnson
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Esmée J van Vliet
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Amber L Zeeman
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Lianne M Wellens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Gerard Llibre-Palomar
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Mario Barrera Román
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Alessia Di Maggio
- Pharmaceutics, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Johanna F Dekkers
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Sabrina Oliveira
- Pharmaceutics, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | | | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Marc Hwa Wijnen
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Ellen J Wehrens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
10
|
Vallance AE. A systematic methodology review of fluorescence-guided cancer surgery to inform the development of a core master protocol and outcome set. BMC Cancer 2024; 24:697. [PMID: 38844894 PMCID: PMC11157717 DOI: 10.1186/s12885-024-12386-4] [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: 01/29/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Fluorescence-guided precision cancer surgery may improve survival and minimize patient morbidity. Efficient development of promising interventions is however hindered by a lack of common methodology. This methodology review aimed to synthesize descriptions of technique, governance processes, surgical learning and outcome reporting in studies of fluorescence-guided cancer surgery to provide guidance for the harmonized design of future studies. METHODS A systematic search of MEDLINE, EMBASE and CENTRAL databases from 2016-2020 identified studies of all designs describing the use of fluorescence in cancer surgery. Dual screening and data extraction was conducted by two independent teams. RESULTS Of 13,108 screened articles, 426 full text articles were included. The number of publications per year increased from 66 in 2016 to 115 in 2020. Indocyanine green was the most commonly used fluorescence agent (391, 91.8%). The most common reported purpose of fluorescence guided surgery was for lymph node mapping (195, 5%) and non-specific tumour visualization (94, 2%). Reporting about surgical learning and governance processes incomplete. A total of 2,577 verbatim outcomes were identified, with the commonly reported outcome lymph node detection (796, 30%). Measures of recurrence (32, 1.2%), change in operative plan (23, 0.9%), health economics (2, 0.1%), learning curve (2, 0.1%) and quality of life (2, 0.1%) were rarely reported. CONCLUSION There was evidence of methodological heterogeneity that may hinder efficient evaluation of fluorescence surgery. Harmonization of the design of future studies may streamline innovation.
Collapse
Affiliation(s)
- Abigail E Vallance
- Centre for Surgical Research, Population Health Sciences, University of Bristol, 39 Whatley Road, Clifton, Bristol, BS8 2PS, UK.
| |
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
|
Roschelle M, Rabbani R, Papageorgiou E, Zhang H, Cooperberg M, Stohr BA, Niknejad A, Anwar M. Multicolor fluorescence microscopy for surgical guidance using a chip-scale imager with a low-NA fiber optic plate and a multi-bandpass interference filter. BIOMEDICAL OPTICS EXPRESS 2024; 15:1761-1776. [PMID: 38495694 PMCID: PMC10942699 DOI: 10.1364/boe.509235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 03/19/2024]
Abstract
In curative-intent cancer surgery, intraoperative fluorescence imaging of both diseased and healthy tissue can help to ensure the successful removal of all gross and microscopic diseases with minimal damage to neighboring critical structures, such as nerves. Current fluorescence-guided surgery (FGS) systems, however, rely on bulky and rigid optics that incur performance-limiting trade-offs between sensitivity and maneuverability. Moreover, many FGS systems are incapable of multiplexed imaging. As a result, clinical FGS is currently limited to millimeter-scale detection of a single fluorescent target. Here, we present a scalable, lens-less fluorescence imaging chip, VISION, capable of sensitive and multiplexed detection within a compact form factor. Central to VISION is a novel optical frontend design combining a low-numerical-aperture fiber optic plate (LNA-FOP) and a multi-bandpass interference filter, which is affixed to a custom CMOS image sensor. The LNA-FOP acts as a planar collimator to improve resolution and compensate for the angle-sensitivity of the interference filter, enabling high-resolution and multiplexed fluorescence imaging without lenses. We show VISION is capable of detecting tumor foci of less than 100 cells at near video framerates and, as proof of principle, can simultaneously visualize both tumors and nerves in ex vivo prostate tissue.
Collapse
Affiliation(s)
- Micah Roschelle
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, USA
| | - Rozhan Rabbani
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, USA
| | - Efthymios Papageorgiou
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, USA
| | - Hui Zhang
- Department of Radiation Oncology, University of California, San Francisco, California 94158, USA
| | - Matthew Cooperberg
- Department of Urology, University of California, San Francisco, California 94158, USA
| | - Bradley A Stohr
- Department of Pathology, University of California, San Francisco, California 94158, USA
| | - Ali Niknejad
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, USA
| | - Mekhail Anwar
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, USA
- Department of Radiation Oncology, University of California, San Francisco, California 94158, USA
| |
Collapse
|
13
|
Pogue BW, Zhu TC, Ntziachristos V, Wilson BC, Paulsen KD, Gioux S, Nordstrom R, Pfefer TJ, Tromberg BJ, Wabnitz H, Yodh A, Chen Y, Litorja M. AAPM Task Group Report 311: Guidance for performance evaluation of fluorescence-guided surgery systems. Med Phys 2024; 51:740-771. [PMID: 38054538 DOI: 10.1002/mp.16849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
The last decade has seen a large growth in fluorescence-guided surgery (FGS) imaging and interventions. With the increasing number of clinical specialties implementing FGS, the range of systems with radically different physical designs, image processing approaches, and performance requirements is expanding. This variety of systems makes it nearly impossible to specify uniform performance goals, yet at the same time, utilization of different devices in new clinical procedures and trials indicates some need for common knowledge bases and a quality assessment paradigm to ensure that effective translation and use occurs. It is feasible to identify key fundamental image quality characteristics and corresponding objective test methods that should be determined such that there are consistent conventions across a variety of FGS devices. This report outlines test methods, tissue simulating phantoms and suggested guidelines, as well as personnel needs and professional knowledge bases that can be established. This report frames the issues with guidance and feedback from related societies and agencies having vested interest in the outcome, coming from an independent scientific group formed from academics and international federal agencies for the establishment of these professional guidelines.
Collapse
Affiliation(s)
- Brian W Pogue
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Timothy C Zhu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vasilis Ntziachristos
- Institute for Biological and Medical Imaging, Technical University of Munich, Helmholtz Zentrum Munich, Munich, Germany
| | - Brian C Wilson
- Department of Medical Biophysics, University of Toronto, University Health Network, Toronto, Ontario, Canada
| | - Keith D Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Sylvain Gioux
- Department of Biomedical Engineering, University of Strasbourg, Strasbourg, France
| | - Robert Nordstrom
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - T Joshua Pfefer
- Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Bruce J Tromberg
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Arjun Yodh
- Department of Physics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yu Chen
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Maritoni Litorja
- Sensor Science Division, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| |
Collapse
|
14
|
Pal R, Lwin TM, Krishnamoorthy M, Collins HR, Chan CD, Prilutskiy A, Nasrallah MP, Dijkhuis TH, Shukla S, Kendall AL, Marshall MS, Carp SA, Hung YP, Shih AR, Martinez-Lage M, Zukerberg L, Sadow PM, Faquin WC, Nahed BV, Feng AL, Emerick KS, Mieog JSD, Vahrmeijer AL, Rajasekaran K, Lee JYK, Rankin KS, Lozano-Calderon S, Varvares MA, Tanabe KK, Kumar ATN. Fluorescence lifetime of injected indocyanine green as a universal marker of solid tumours in patients. Nat Biomed Eng 2023; 7:1649-1666. [PMID: 37845517 DOI: 10.1038/s41551-023-01105-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 09/10/2023] [Indexed: 10/18/2023]
Abstract
The surgical resection of solid tumours can be enhanced by fluorescence-guided imaging. However, variable tumour uptake and incomplete clearance of fluorescent dyes reduces the accuracy of distinguishing tumour from normal tissue via conventional fluorescence intensity-based imaging. Here we show that, after systemic injection of the near-infrared dye indocyanine green in patients with various types of solid tumour, the fluorescence lifetime (FLT) of tumour tissue is longer than the FLT of non-cancerous tissue. This tumour-specific shift in FLT can be used to distinguish tumours from normal tissue with an accuracy of over 97% across tumour types, and can be visualized at the cellular level using microscopy and in larger specimens through wide-field imaging. Unlike fluorescence intensity, which depends on imaging-system parameters, tissue depth and the amount of dye taken up by tumours, FLT is a photophysical property that is largely independent of these factors. FLT imaging with indocyanine green may improve the accuracy of cancer surgeries.
Collapse
Affiliation(s)
- Rahul Pal
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Thinzar M Lwin
- Department of Surgical Oncology, City of Hope Hospital, Duarte, CA, USA
| | - Murali Krishnamoorthy
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hannah R Collins
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Corey D Chan
- Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andrey Prilutskiy
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - MacLean P Nasrallah
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Tom H Dijkhuis
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Shriya Shukla
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Amy L Kendall
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Michael S Marshall
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefan A Carp
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yin P Hung
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Angela R Shih
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Martinez-Lage
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lawrence Zukerberg
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology and Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - William C Faquin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Otolaryngology and Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Brian V Nahed
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Allen L Feng
- Department of Otolaryngology and Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Kevin S Emerick
- Department of Otolaryngology and Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Karthik Rajasekaran
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth S Rankin
- The North of England Bone and Soft Tissue Tumour Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Santiago Lozano-Calderon
- Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark A Varvares
- Department of Otolaryngology and Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Kenneth K Tanabe
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand T N Kumar
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
15
|
Tanniche I, Behkam B. Engineered live bacteria as disease detection and diagnosis tools. J Biol Eng 2023; 17:65. [PMID: 37875910 PMCID: PMC10598922 DOI: 10.1186/s13036-023-00379-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
Sensitive and minimally invasive medical diagnostics are essential to the early detection of diseases, monitoring their progression and response to treatment. Engineered bacteria as live sensors are being developed as a new class of biosensors for sensitive, robust, noninvasive, and in situ detection of disease onset at low cost. Akin to microrobotic systems, a combination of simple genetic rules, basic logic gates, and complex synthetic bioengineering principles are used to program bacterial vectors as living machines for detecting biomarkers of diseases, some of which cannot be detected with other sensing technologies. Bacterial whole-cell biosensors (BWCBs) can have wide-ranging functions from detection only, to detection and recording, to closed-loop detection-regulated treatment. In this review article, we first summarize the unique benefits of bacteria as living sensors. We then describe the different bacteria-based diagnosis approaches and provide examples of diagnosing various diseases and disorders. We also discuss the use of bacteria as imaging vectors for disease detection and image-guided surgery. We conclude by highlighting current challenges and opportunities for further exploration toward clinical translation of these bacteria-based systems.
Collapse
Affiliation(s)
- Imen Tanniche
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
- School of Biomedical Engineered and Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
- Center for Engineered Health, Institute for Critical Technology and Applied Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
16
|
Kennedy GT, Azari FS, Chang A, Chang A, Bou-Samra P, Desphande C, Delikatny EJ, Eruslanov E, Kucharczuk JC, Rice DC, Singhal S. A pH-Activatable Nanoprobe Labels Diverse Histologic Subtypes of Human Lung Cancer During Resection. Mol Imaging Biol 2023; 25:824-832. [PMID: 37697109 PMCID: PMC11141135 DOI: 10.1007/s11307-023-01853-2] [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: 05/01/2023] [Revised: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND Intraoperative molecular imaging (IMI) uses tumor-targeted optical contrast agents to improve identification and clearance of cancer during surgery. Recently, pH-activatable contrast agents have been developed but none has been tested in lung cancer. Here, we report the successful clinical translation of pegsitacianine (ONM-100), a pH-activatable nanoprobe, for fluorescence-guided lung cancer resection. METHODS We first characterized the pH setpoint for pegsitacianine fluorescence activation in vitro. We then optimized the specificity, dosing, and timing of pegsitacianine in murine flank xenograft models of lung adenocarcinoma and squamous cell carcinoma. Finally, we tested pegsitacianine in humans undergoing lung cancer surgery as part of an ongoing phase 2 trial. RESULTS We found that the fluorescence activation of pegsitacianine occurred below physiologic pH in vitro. Using preclinical models of lung cancer, we found that the probe selectively labeled both adenocarcinoma and squamous cell carcinoma xenografts (mean tumor-to-background ratio [TBR] > 2.0 for all cell lines). In the human pilot study, we report cases in which pegsitacianine localized pulmonary adenocarcinoma and pulmonary squamous cell carcinoma (TBRs= 2.7 and 2.4) in real time to illustrate its successful clinical translation and potential to improve surgical management. CONCLUSIONS This translational study demonstrates the feasibility of pegsitacianine as an IMI probe to label the two most common histologic subtypes of human lung cancer.
Collapse
Affiliation(s)
- Gregory Thomas Kennedy
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard, 14th Floor, South Pavilion, Philadelphia, PA, 19104, USA
| | - Feredun S Azari
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard, 14th Floor, South Pavilion, Philadelphia, PA, 19104, USA
| | - Ashley Chang
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard, 14th Floor, South Pavilion, Philadelphia, PA, 19104, USA
| | - Austin Chang
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard, 14th Floor, South Pavilion, Philadelphia, PA, 19104, USA
| | - Patrick Bou-Samra
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard, 14th Floor, South Pavilion, Philadelphia, PA, 19104, USA
| | - Charuhas Desphande
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Edward J Delikatny
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Evgeniy Eruslanov
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard, 14th Floor, South Pavilion, Philadelphia, PA, 19104, USA
| | - John C Kucharczuk
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard, 14th Floor, South Pavilion, Philadelphia, PA, 19104, USA
| | - David C Rice
- Department of Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania School of Medicine, 3400 Civic Center Boulevard, 14th Floor, South Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
17
|
Ochoa MI, Ruiz A, LaRochelle E, Reed M, Berber E, Poultsides G, Pogue BW. Assessment of open-field fluorescence guided surgery systems: implementing a standardized method for characterization and comparison. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:096007. [PMID: 37745774 PMCID: PMC10513724 DOI: 10.1117/1.jbo.28.9.096007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/26/2023]
Abstract
Significance Fluorescence guided surgery (FGS) has demonstrated improvements in decision making and patient outcomes for a wide range of surgical procedures. Not only can FGS systems provide a higher level of structural perfusion accuracy in tissue reconstruction cases but they can also serve for real-time functional characterization. Multiple FGS devices have been Food and Drug administration (FDA) cleared for use in open and laparoscopic surgery. Despite the rapid growth of the field, there has been a lack standardization methods. Aim This work overviews commonalities inherent to optical imaging methods that can be exploited to produce such a standardization procedure. Furthermore, a system evaluation pipeline is proposed and executed through the use of photo-stable indocyanine green fluorescence phantoms. Five different FDA-approved open-field FGS systems are used and evaluated with the proposed method. Approach The proposed pipeline encompasses the following characterization: (1) imaging spatial resolution and sharpness, (2) sensitivity and linearity, (3) imaging depth into tissue, (4) imaging system DOF, (5) uniformity of illumination, (6) spatial distortion, (7) signal to background ratio, (8) excitation bands, and (9) illumination wavelength and power. Results The results highlight how such a standardization approach can be successfully implemented for inter-system comparisons as well as how to better understand essential features within each FGS setup. Conclusions Despite clinical use being the end goal, a robust yet simple standardization pipeline before clinical trials, such as the one presented herein, should benefit regulatory agencies, manufacturers, and end-users to better assess basic performance and improvements to be made in next generation FGS systems.
Collapse
Affiliation(s)
- Marien I. Ochoa
- University of Wisconsin Madison, Department of Medical Physics, Madison, Wisconsin, United States
| | - Alberto Ruiz
- QUEL Imaging, White River Junction, Vermont, United States
| | | | - Matthew Reed
- University of Wisconsin Madison, Department of Medical Physics, Madison, Wisconsin, United States
| | - Eren Berber
- Cleveland Clinic - Marymount Hospital, Garfield Heights, Ohio, United States
| | - George Poultsides
- Stanford Medicine, Department of Surgery, Stanford, California, United States
| | - Brian W. Pogue
- University of Wisconsin Madison, Department of Medical Physics, Madison, Wisconsin, United States
| |
Collapse
|
18
|
Srivastava I, Lew B, Wang Y, Blair S, George MB, Hajek BS, Bangru S, Pandit S, Wang Z, Ludwig J, Flatt K, Gruebele M, Nie S, Gruev V. Cell-Membrane Coated Nanoparticles for Tumor Delineation and Qualitative Estimation of Cancer Biomarkers at Single Wavelength Excitation in Murine and Phantom Models. ACS NANO 2023; 17:8465-8482. [PMID: 37126072 DOI: 10.1021/acsnano.3c00578] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Real-time guidance through fluorescence imaging improves the surgical outcomes of tumor resections, reducing the chances of leaving positive margins behind. As tumors are heterogeneous, it is imperative to interrogate multiple overexpressed cancer biomarkers with high sensitivity and specificity to improve surgical outcomes. However, for accurate tumor delineation and ratiometric detection of tumor biomarkers, current methods require multiple excitation wavelengths to image multiple biomarkers, which is impractical in a clinical setting. Here, we have developed a biomimetic platform comprising near-infrared fluorescent semiconducting polymer nanoparticles (SPNs) with red blood cell membrane (RBC) coating, capable of targeting two representative cell-surface biomarkers (folate, αυβ3 integrins) using a single excitation wavelength for tumor delineation during surgical interventions. We evaluate our single excitation ratiometric nanoparticles in in vitro tumor cells, ex vivo tumor-mimicking phantoms, and in vivo mouse xenograft tumor models. Favorable biological properties (improved biocompatibility, prolonged blood circulation, reduced liver uptake) are complemented by superior spectral features: (i) specific fluorescence enhancement in tumor regions with high tumor-to-normal tissue (T/NT) ratios in ex vivo samples and (ii) estimation of cell-surface tumor biomarkers with single wavelength excitation providing insights about cancer progression (metastases). Our single excitation, dual output approach has the potential to differentiate between the tumor and healthy regions and simultaneously provide a qualitative indicator of cancer progression, thereby guiding surgeons in the operating room with the resection process.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sushant Bangru
- Department of Cell Biology, Duke University, Durham, North Carolina 27705, United States
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Price LJ, Tatz J, Sutin J, Spring BQ. Multi-objective optimization of custom compound prism arrays for multiplexed optical imaging. OPTICS EXPRESS 2023; 31:9739-9749. [PMID: 37157537 PMCID: PMC10316679 DOI: 10.1364/oe.475175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/06/2022] [Accepted: 12/11/2022] [Indexed: 05/10/2023]
Abstract
Compound prism arrays are a powerful, yet underutilized, solution for producing high transmission and customized chromatic dispersion profiles over broad bandwidths, the quality of which is unobtainable with commercially available prisms or diffraction gratings. However, the computational complexity associated with designing these prism arrays presents a barrier to the widespread adoption of their use. Here we introduce customizable prism designer software that facilitates high-speed optimization of compound arrays guided by target specifications for chromatic dispersion linearity and detector geometry. Information theory is utilized such that target parameters can be easily modified through user input to efficiently simulate a broad range of possible prism array designs. We demonstrate the capabilities of the designer software to simulate new prism array designs for multiplexed, hyperspectral microscopy that achieve chromatic dispersion linearity and a 70-90% light transmission over a significant portion of the visible wavelength range (500-820 nm). The designer software is applicable to many optical spectroscopy and spectral microscopy applications-with varying requirements for spectral resolution, light ray deviation, and physical size-that are photon-starved and for which the enhanced transmission of refraction versus diffraction warrants custom optical designs.
Collapse
Affiliation(s)
- Liam J. Price
- Department of Physics, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Julia Tatz
- Department of Physics, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
- Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jason Sutin
- Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Bryan Q. Spring
- Department of Physics, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
- Department of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| |
Collapse
|
20
|
Streeter SS, Ray GS, Bateman LM, Hebert KA, Bushee FE, Rodi SW, Gitajn IL, Ahn J, Singhal S, Martin ND, Bernthal NM, Lee C, Obremskey WT, Schoenecker JG, Elliott JT, Henderson ER. Early identification of life-threatening soft-tissue infection using dynamic fluorescence imaging: first-in-kind clinical study of first-pass kinetics. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2023; 12361:123610B. [PMID: 37034555 PMCID: PMC10078977 DOI: 10.1117/12.2648408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
Necrotizing soft-tissue infections (NSTIs) are aggressive and deadly. Immediate surgical debridement is standard-of-care, but patients often present with non-specific symptoms, thereby delaying treatment. Because NSTIs cause microvascular thrombosis, we hypothesized that perfusion imaging using indocyanine green (ICG) would show diminished fluorescence signal in NSTI-affected tissues, particularly compared to non-necrotizing, superficial infections. Through a first-in-kind clinical study, we performed first-pass ICG fluorescence perfusion imaging of patients with suspected NSTIs. Early results support our hypothesis that ICG signal voids occur in NSTI-affected tissues and that dynamic contrast-enhanced fluorescence parameters reveal tissue kinetics that may be related to disease progression and extent.
Collapse
Affiliation(s)
- Samuel S. Streeter
- Department of Orthopaedics, Dartmouth Health, Lebanon, NH 03756
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755
| | - Gabrielle S. Ray
- Department of Orthopaedics, Dartmouth Health, Lebanon, NH 03756
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755
| | - Logan M. Bateman
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755
| | - Kendra A. Hebert
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755
| | | | - Scott W. Rodi
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755
| | - I. Leah Gitajn
- Department of Orthopaedics, Dartmouth Health, Lebanon, NH 03756
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755
| | - Jaimo Ahn
- Michigan Medicine, U. of Michigan, Ann Arbor, MI 48109
| | - Sunil Singhal
- Perelman School of Medicine, U. of Pennsylvania, Philadelphia, PA 19104
| | - Niels D. Martin
- Perelman School of Medicine, U. of Pennsylvania, Philadelphia, PA 19104
| | - Nicholas M. Bernthal
- David Geffen School of Medicine, U. of California Los Angeles, Santa Monica, CA 90404
| | - Christopher Lee
- David Geffen School of Medicine, U. of California Los Angeles, Santa Monica, CA 90404
| | | | | | - Jonathan Thomas Elliott
- Department of Orthopaedics, Dartmouth Health, Lebanon, NH 03756
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755
| | - Eric R. Henderson
- Department of Orthopaedics, Dartmouth Health, Lebanon, NH 03756
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755
| |
Collapse
|
21
|
Hernandez Vargas S, AghaAmiri S, Ghosh SC, Luciano MP, Borbon LC, Ear PH, Howe JR, Bailey-Lundberg JM, Simonek GD, Halperin DM, Tran Cao HS, Ikoma N, Schnermann MJ, Azhdarinia A. High-Contrast Detection of Somatostatin Receptor Subtype-2 for Fluorescence-Guided Surgery. Mol Pharm 2022; 19:4241-4253. [PMID: 36174110 PMCID: PMC9830638 DOI: 10.1021/acs.molpharmaceut.2c00583] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Dye design can influence the ability of fluorescently labeled imaging agents to generate tumor contrast and has become an area of significant interest in the field of fluorescence-guided surgery (FGS). Here, we show that the charge-balanced near-infrared fluorescent (NIRF) dye FNIR-Tag enhances the imaging properties of a fluorescently labeled somatostatin analogue. In vitro studies showed that the optimized fluorescent conjugate MMC(FNIR-Tag)-TOC bound primarily via somatostatin receptor subtype-2 (SSTR2), whereas its negatively charged counterpart with IRDye 800CW had higher off-target binding. NIRF imaging in cell line- and patient-derived xenograft models revealed markedly higher tumor contrast with MMC(FNIR-Tag)-TOC, which was attributed to increased tumor specificity. Ex vivo staining of surgical biospecimens from primary and metastatic tumors, as well as involved lymph nodes, demonstrated binding to human tumors. Finally, in an orthotopic tumor model, a simulated clinical workflow highlighted our unique ability to use standard preoperative nuclear imaging for selecting patients likely to benefit from SSTR2-targeted FGS. Our findings demonstrate the translational potential of MMC(FNIR-Tag)-TOC for intraoperative imaging and suggest broad utility for using FNIR-Tag in fluorescent probe development.
Collapse
Affiliation(s)
- Servando Hernandez Vargas
- The
Brown Foundation Institute of Molecular Medicine, McGovern Medical
School, The University of Texas Health Science
Center at Houston, Houston, Texas77054, United States
| | - Solmaz AghaAmiri
- The
Brown Foundation Institute of Molecular Medicine, McGovern Medical
School, The University of Texas Health Science
Center at Houston, Houston, Texas77054, United States
| | - Sukhen C. Ghosh
- The
Brown Foundation Institute of Molecular Medicine, McGovern Medical
School, The University of Texas Health Science
Center at Houston, Houston, Texas77054, United States
| | - Michael P. Luciano
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland21702, United States
| | - Luis C. Borbon
- Department
of Surgery, University of Iowa Carver College
of Medicine, Iowa City, Iowa52242, United States
| | - Po Hien Ear
- Department
of Surgery, University of Iowa Carver College
of Medicine, Iowa City, Iowa52242, United States
| | - James R. Howe
- Department
of Surgery, University of Iowa Carver College
of Medicine, Iowa City, Iowa52242, United States
| | - Jennifer M. Bailey-Lundberg
- Department
of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas77030, United States
| | - Gregory D. Simonek
- Center
for Laboratory Animal Medicine and Care, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas77030, United States
| | - Daniel M. Halperin
- Department
of Gastrointestinal Medical Oncology, The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, Texas77030, United States
| | - Hop S. Tran Cao
- Department
of Surgical Oncology, The University of
Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, Texas77030, United
States
| | - Naruhiko Ikoma
- Department
of Surgical Oncology, The University of
Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, Texas77030, United
States
| | - Martin J. Schnermann
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland21702, United States
| | - Ali Azhdarinia
- The
Brown Foundation Institute of Molecular Medicine, McGovern Medical
School, The University of Texas Health Science
Center at Houston, Houston, Texas77054, United States,
| |
Collapse
|
22
|
Olson MT, Aguilar EN, Brooks CL, Isder CC, Muilenburg KM, Talmon GA, Ly QP, Carlson MA, Hollingsworth MA, Mohs AM. Preclinical Evaluation of a Humanized, Near-Infrared Fluorescent Antibody for Fluorescence-Guided Surgery of MUC16-Expressing Pancreatic Cancer. Mol Pharm 2022; 19:3586-3599. [PMID: 35640060 PMCID: PMC9864431 DOI: 10.1021/acs.molpharmaceut.2c00203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Surgery remains the only potentially curative treatment option for pancreatic cancer, but resections are made more difficult by infiltrative disease, proximity of critical vasculature, peritumoral inflammation, and dense stroma. Surgeons are limited to tactile and visual cues to differentiate cancerous tissue from normal tissue. Furthermore, translating preoperative images to the intraoperative setting poses additional challenges for tumor detection, and can result in undetected and unresected lesions. Thus, pancreatic ductal adenocarcinoma (PDAC) has high rates of incomplete resections, and subsequently, disease recurrence. Fluorescence-guided surgery (FGS) has emerged as a method to improve intraoperative detection of cancer and ultimately improve surgical outcomes. Initial clinical trials have demonstrated feasibility of FGS for PDAC, but there are limited targeted probes under investigation for this disease, highlighting the need for development of additional novel biomarkers to reflect the PDAC heterogeneity. MUCIN16 (MUC16) is a glycoprotein that is overexpressed in 60-80% of PDAC. In our previous work, we developed a MUC16-targeted murine antibody near-infrared conjugate, termed AR9.6-IRDye800, that showed efficacy in detecting pancreatic cancer. To build on the translational potential of this imaging probe, a humanized variant of the AR9.6 fluorescent conjugate was developed and investigated herein. This conjugate, termed huAR9.6-IRDye800, showed equivalent binding properties to its murine counterpart. Using an optimized dye:protein ratio of 1:1, in vivo studies demonstrated high tumor to background ratios in MUC16-expressing tumor models, and delineation of tumors in a patient-derived xenograft model. Safety, biodistribution, and toxicity studies were conducted. These studies demonstrated that huAR9.6-IRDye800 was safe, did not yield evidence of histological toxicity, and was well tolerated in vivo. The results from this work suggest that AR9.6-IRDye800 is an efficacious and safe imaging agent for identifying pancreatic cancer intraoperatively through fluorescence-guided surgery.
Collapse
Affiliation(s)
- Madeline T. Olson
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Eric N. Aguilar
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA 93740
| | - Cory L. Brooks
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA 93740
| | - Carly C. Isder
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Kathtyn M. Muilenburg
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
| | - Geoffrey A. Talmon
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Quan P. Ly
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198
| | - Mark A. Carlson
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Surgery, VA Medical Center, Omaha, NE 68105
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Aaron M. Mohs
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
23
|
Blair S, Garcia M, Zhu Z, Liang Z, Lew B, George M, Kondov B, Stojanoski S, Todorovska MB, Miladinova D, Kondov G, Gruev V. Decoupling channel count from field of view and spatial resolution in single-sensor imaging systems for fluorescence image-guided surgery. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-220139GR. [PMID: 36163641 PMCID: PMC9511017 DOI: 10.1117/1.jbo.27.9.096006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/07/2022] [Indexed: 05/17/2023]
Abstract
Significance Near-infrared fluorescence image-guided surgery is often thought of as a spectral imaging problem where the channel count is the critical parameter, but it should also be thought of as a multiscale imaging problem where the field of view and spatial resolution are similarly important. Aim Conventional imaging systems based on division-of-focal-plane architectures suffer from a strict relationship between the channel count on one hand and the field of view and spatial resolution on the other, but bioinspired imaging systems that combine stacked photodiode image sensors and long-pass/short-pass filter arrays offer a weaker tradeoff. Approach In this paper, we explore how the relevant changes to the image sensor and associated image processing routines affect image fidelity during image-guided surgeries for tumor removal in an animal model of breast cancer and nodal mapping in women with breast cancer. Results We demonstrate that a transition from a conventional imaging system to a bioinspired one, along with optimization of the image processing routines, yields improvements in multiple measures of spectral and textural rendition relevant to surgical decision-making. Conclusions These results call for a critical examination of the devices and algorithms that underpin image-guided surgery to ensure that surgeons receive high-quality guidance and patients receive high-quality outcomes as these technologies enter clinical practice.
Collapse
Affiliation(s)
- Steven Blair
- University of Illinois at Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
| | - Missael Garcia
- University of Illinois at Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
| | - Zhongmin Zhu
- University of Illinois at Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
| | - Zuodong Liang
- University of Illinois at Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
| | - Benjamin Lew
- University of Illinois at Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
| | - Mebin George
- University of Illinois at Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
| | - Borislav Kondov
- University Clinic Hospital, Ss. Cyril and Methodius University of Skopje, Department of Thoracic and Vascular Surgery, Skopje, Republic of North Macedonia
| | - Sinisa Stojanoski
- University Clinic Hospital, Ss. Cyril and Methodius University of Skopje, Institute of Pathophysiology and Nuclear Medicine, Skopje, Republic of North Macedonia
| | - Magdalena Bogdanovska Todorovska
- University Clinic Hospital, Ss. Cyril and Methodius University of Skopje, Department of Pathology, Skopje, Republic of North Macedonia
| | - Daniela Miladinova
- University Clinic Hospital, Ss. Cyril and Methodius University of Skopje, Institute of Pathophysiology and Nuclear Medicine, Skopje, Republic of North Macedonia
| | - Goran Kondov
- University Clinic Hospital, Ss. Cyril and Methodius University of Skopje, Department of Thoracic and Vascular Surgery, Skopje, Republic of North Macedonia
| | - Viktor Gruev
- University of Illinois at Urbana-Champaign, Department of Electrical and Computer Engineering, Urbana, Illinois, United States
- University of Illinois at Urbana-Champaign, Beckman Institute for Advanced Science and Technology, Urbana, Illinois, United States
- University of Illinois at Urbana-Champaign, Carle Illinois College of Medicine, Urbana, Illinois, United States
| |
Collapse
|
24
|
Harman RC, Lang RT, Kercher EM, Leven P, Spring BQ. Denoising multiplexed microscopy images in n-dimensional spectral space. BIOMEDICAL OPTICS EXPRESS 2022; 13:4298-4309. [PMID: 36032573 PMCID: PMC9408246 DOI: 10.1364/boe.463979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 06/15/2023]
Abstract
Hyperspectral fluorescence microscopy images of biological specimens frequently contain multiple observations of a sparse set of spectral features spread in space with varying intensity. Here, we introduce a spectral vector denoising algorithm that filters out noise without sacrificing spatial information by leveraging redundant observations of spectral signatures. The algorithm applies an n-dimensional Chebyshev or Fourier transform to cluster pixels based on spectral similarity independent of pixel intensity or location, and a denoising convolution filter is then applied in this spectral space. The denoised image may then undergo spectral decomposition analysis with enhanced accuracy. Tests utilizing both simulated and empirical microscopy data indicate that denoising in 3 to 5-dimensional (3D to 5D) spectral spaces decreases unmixing error by up to 70% without degrading spatial resolution.
Collapse
Affiliation(s)
- Rebecca C. Harman
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
- Department of Physics, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
| | - Ryan T. Lang
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
- Department of Physics, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
| | - Eric M. Kercher
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
- Department of Physics, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
| | - Paige Leven
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
- Department of Physics, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
| | - Bryan Q. Spring
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
- Department of Physics, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA 02115, USA
| |
Collapse
|
25
|
Lwin TM, Turner MA, Nishino H, Amirfakhri S, Hernot S, Hoffman RM, Bouvet M. Fluorescent Anti-CEA Nanobody for Rapid Tumor-Targeting and Imaging in Mouse Models of Pancreatic Cancer. Biomolecules 2022; 12:711. [PMID: 35625638 PMCID: PMC9138244 DOI: 10.3390/biom12050711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 12/16/2022] Open
Abstract
Tumor-specific targeting with fluorescent probes can enhance contrast for identification of cancer during surgical resection and visualize otherwise invisible tumor margins. Nanobodies are the smallest naturally-occurring antigen-binding molecules with rapid pharmacokinetics. The present work demonstrates the efficacy of a fluorescent anti-CEA nanobody conjugated to an IR800 dye to target and label patient derived pancreatic cancer xenografts. After intravenous administration, the probe rapidly localized to the pancreatic cancer tumors within an hour and had a tumor-to-background ratio of 2.0 by 3 h. The fluorescence signal was durable over a prolonged period of time. With the rapid kinetics afforded by fluorescent nanobodies, both targeting and imaging can be performed on the same day as surgery.
Collapse
Affiliation(s)
- Thinzar M. Lwin
- Department of Surgery, University of California San Diego, San Diego, CA 92093, USA; (T.M.L.); (M.A.T.); (H.N.); (S.A.); (R.M.H.)
- Department of Surgical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Michael A. Turner
- Department of Surgery, University of California San Diego, San Diego, CA 92093, USA; (T.M.L.); (M.A.T.); (H.N.); (S.A.); (R.M.H.)
- Department of Surgery, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Hiroto Nishino
- Department of Surgery, University of California San Diego, San Diego, CA 92093, USA; (T.M.L.); (M.A.T.); (H.N.); (S.A.); (R.M.H.)
- Department of Surgery, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, San Diego, CA 92093, USA; (T.M.L.); (M.A.T.); (H.N.); (S.A.); (R.M.H.)
- Department of Surgery, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Sophie Hernot
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, B-1090 Brussels, Belgium;
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, San Diego, CA 92093, USA; (T.M.L.); (M.A.T.); (H.N.); (S.A.); (R.M.H.)
- Department of Surgery, VA San Diego Healthcare System, San Diego, CA 92161, USA
- AntiCancer, Inc., San Diego, CA 92111, USA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, CA 92093, USA; (T.M.L.); (M.A.T.); (H.N.); (S.A.); (R.M.H.)
- Department of Surgery, VA San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
26
|
Hacker L, Wabnitz H, Pifferi A, Pfefer TJ, Pogue BW, Bohndiek SE. Criteria for the design of tissue-mimicking phantoms for the standardization of biophotonic instrumentation. Nat Biomed Eng 2022; 6:541-558. [PMID: 35624150 DOI: 10.1038/s41551-022-00890-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/07/2022] [Indexed: 01/08/2023]
Abstract
A lack of accepted standards and standardized phantoms suitable for the technical validation of biophotonic instrumentation hinders the reliability and reproducibility of its experimental outputs. In this Perspective, we discuss general criteria for the design of tissue-mimicking biophotonic phantoms, and use these criteria and state-of-the-art developments to critically review the literature on phantom materials and on the fabrication of phantoms. By focusing on representative examples of standardization in diffuse optical imaging and spectroscopy, fluorescence-guided surgery and photoacoustic imaging, we identify unmet needs in the development of phantoms and a set of criteria (leveraging characterization, collaboration, communication and commitment) for the standardization of biophotonic instrumentation.
Collapse
Affiliation(s)
- Lina Hacker
- Department of Physics, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Heidrun Wabnitz
- Physikalisch-Technische Bundesanstalt (PTB), Berlin, Germany
| | | | | | - Brian W Pogue
- Thayer School of Engineering, Dartmouth, Hanover, NH, USA
| | - Sarah E Bohndiek
- Department of Physics, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Fluorescence Molecular Targeting of Colon Cancer to Visualize the Invisible. Cells 2022; 11:cells11020249. [PMID: 35053365 PMCID: PMC8773892 DOI: 10.3390/cells11020249] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/28/2021] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) is a common cause of cancer and cancer-related death. Surgery is the only curative modality. Fluorescence-enhanced visualization of CRC with targeted fluorescent probes that can delineate boundaries and target tumor-specific biomarkers can increase rates of curative resection. Approaches to enhancing visualization of the tumor-to-normal tissue interface are active areas of investigation. Nonspecific dyes are the most-used approach, but tumor-specific targeting agents are progressing in clinical trials. The present narrative review describes the principles of fluorescence targeting of CRC for diagnosis and fluorescence-guided surgery with molecular biomarkers for preclinical or clinical evaluation.
Collapse
|
28
|
Mendez CB, Gonda A, Shah JV, Siebert JN, Zhao X, He S, Riman RE, Tan MC, Moghe PV, Ganapathy V, Pierce MC. Short-Wave Infrared Emitting Nanocomposites for Fluorescence-Guided Surgery. IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS : A PUBLICATION OF THE IEEE LASERS AND ELECTRO-OPTICS SOCIETY 2021; 27:7300307. [PMID: 36710719 PMCID: PMC9881055 DOI: 10.1109/jstqe.2021.3066895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Fluorescence-guided surgery (FGS) is an emerging technique for tissue visualization during surgical procedures. Structures of interest are labeled with exogenous probes whose fluorescent emissions are acquired and viewed in real-time with optical imaging systems. This study investigated rare-earth-doped albumin-encapsulated nanocomposites (REANCs) as short-wave infrared emitting contrast agents for FGS. Experiments were conducted using an animal model of 4T1 breast cancer. The signal-to-background ratio (SBR) obtained with REANCs was compared to values obtained using indocyanine green (ICG), a near-infrared dye used in clinical practice. Prior to resection, the SBR for tumors following intratumoral administration of REANCs was significantly higher than for tumors injected with ICG. Following FGS, evaluation of fluorescence intensity levels in excised tumors and at the surgical bed demonstrated higher contrast between tissues at these sites with REANC contrast than ICG. REANCs also demonstrated excellent photostability over 2 hours of continuous illumination, as well as the ability to perform FGS under ambient lighting, establishing these nanocomposites as a promising contrast agent for FGS applications.
Collapse
Affiliation(s)
- Carolina Bobadilla Mendez
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Amber Gonda
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Jay V Shah
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Jake N Siebert
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Xinyu Zhao
- Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Shuqing He
- Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Richard E Riman
- Department of Materials Science and Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Mei Chee Tan
- Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Prabhas V Moghe
- Department of Biomedical Engineering, and the Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Vidya Ganapathy
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Mark C Pierce
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| |
Collapse
|
29
|
Restall BS, Kedarisetti P, Haven NJM, Martell MT, Zemp RJ. Multimodal 3D photoacoustic remote sensing and confocal fluorescence microscopy imaging. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-210059R. [PMID: 34523269 PMCID: PMC8440567 DOI: 10.1117/1.jbo.26.9.096501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 08/06/2021] [Indexed: 05/25/2023]
Abstract
SIGNIFICANCE Complementary absorption and fluorescence contrast could prove useful for a wide range of biomedical applications. However, current absorption-based photoacoustic microscopy systems require the ultrasound transducers to physically touch the samples, thereby increasing contamination and limiting strong optical focusing in reflection mode. AIM We sought to develop an all-optical system for imaging cells and tissues using the three combined imaging modalities: photoacoustic remote sensing (PARS), epifluorescence, and confocal laser scanning microscopy (CLSM). APPROACH A PARS subsystem with ultraviolet excitation was used to obtain label-free absorption-contrast images of nucleic acids in ex vivo tissue samples. Co-integrated epifluorescence and CLSM subsystems were used to verify the 2D and 3D nuclei distribution. RESULTS Complementary absorption and fluorescence contrast were demonstrated in phantom imaging experiments and subsequent cell and tissue imaging experiments. Lateral and axial resolution of ultraviolet-PARS (UV-PARS) is shown to be 0.39 and 1.6 μm, respectively, with 266-nm light. CLSM lateral and axial resolution was measured as 0.97 and 2.0 μm, respectively. This resolution is sufficient to image individual cell layers with fine optical sectioning. UV-PARS images of cell nuclei are validated in thick tissue using CLSM. CONCLUSIONS Multimodal absorption and fluorescence contrast are obtained with a non-contact all-optical microscopy system for the first time and utilized to obtain images of cells and tissues with subcellular resolution.
Collapse
Affiliation(s)
- Brendon S. Restall
- University of Alberta, Department of Electrical and Computer Engineering, Edmonton, Canada
| | - Pradyumna Kedarisetti
- University of Alberta, Department of Electrical and Computer Engineering, Edmonton, Canada
| | - Nathaniel J. M. Haven
- University of Alberta, Department of Electrical and Computer Engineering, Edmonton, Canada
| | - Matthew T. Martell
- University of Alberta, Department of Electrical and Computer Engineering, Edmonton, Canada
| | - Roger J. Zemp
- University of Alberta, Department of Electrical and Computer Engineering, Edmonton, Canada
| |
Collapse
|
30
|
Stewart HL, Birch DJS. Fluorescence Guided Surgery. Methods Appl Fluoresc 2021; 9. [PMID: 34399409 DOI: 10.1088/2050-6120/ac1dbb] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/16/2021] [Indexed: 01/22/2023]
Abstract
Fluorescence guided surgery (FGS) is an imaging technique that allows the surgeon to visualise different structures and types of tissue during a surgical procedure that may not be as visible under white light conditions. Due to the many potential advantages of fluorescence guided surgery compared to more traditional clinical imaging techniques such as its higher contrast and sensitivity, less subjective use, and ease of instrument operation, the research interest in fluorescence guided surgery continues to grow over various key aspects such as fluorescent probe development and surgical system development as well as its potential clinical applications. This review looks to summarise some of the emerging opportunities and developments that have already been made in fluorescence guided surgery in recent years while highlighting its advantages as well as limitations that need to be overcome in order to utilise the full potential of fluorescence within the surgical environment.
Collapse
Affiliation(s)
- Hazel L Stewart
- Translational Healthcare Technologies Group, Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - David J S Birch
- Department of Physics, The Photophysics Research Group, University of Strathclyde, SUPA, John Anderson Building, 107 Rottenrow East, Glasgow G4 0NG, United Kingdom
| |
Collapse
|
31
|
Debacker JM, Schelfhout V, Brochez L, Creytens D, D’Asseler Y, Deron P, Keereman V, Van de Vijver K, Vanhove C, Huvenne W. High-Resolution 18F-FDG PET/CT for Assessing Three-Dimensional Intraoperative Margins Status in Malignancies of the Head and Neck, a Proof-of-Concept. J Clin Med 2021; 10:jcm10163737. [PMID: 34442033 PMCID: PMC8397229 DOI: 10.3390/jcm10163737] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 01/27/2023] Open
Abstract
The surgical treatment of head and neck malignancies relies on the complete removal of tumoral tissue, while inadequate margins necessitate the use of adjuvant therapy. However, most positive margins are identified postoperatively as deep margins, and intraoperative identification of the deep positive margins could help achieve adequate surgical margins and decrease adjuvant therapies. To improve deep-margin identification, we investigated whether the use of high-resolution preclinical PET and CT could increase certainty about the surgical margins in three dimensions. Patients with a malignancy of the head and neck planned for surgical resection were administered a clinical activity of 4MBq/kg 18F-FDG approximately one hour prior to surgical initiation. Subsequently, the resected specimen was scanned with a micro-PET-CT imaging device, followed by histopathological assessment. Eight patients were included in the study and intraoperative PET/CT-imaging of 11 tumoral specimens and lymph nodes of three patients was performed. As a result of the increased resolution, differentiation between inflamed and dysplastic tissue versus malignant tissue was complicated in malignancies with increased peritumoral inflammation. The current technique allowed the three-dimensional delineation of 18F-FDG using submillimetric PET/CT imaging. While further optimization and patient stratification is required, clinical implementation could enable deep margin assessment in head and neck resection specimens.
Collapse
Affiliation(s)
- Jens M. Debacker
- Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (L.B.); (P.D.); (W.H.)
- Department of Head and Neck Surgery, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Nuclear Medicine, University Hospital Brussels, 1090 Brussels, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
- Correspondence: ; Tel.: +32-9-332-39-90
| | - Vanessa Schelfhout
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lieve Brochez
- Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (L.B.); (P.D.); (W.H.)
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
- Department of Dermatology, Ghent University Hospital, 9000 Ghent, Belgium
| | - David Creytens
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
- Department of Pathology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Yves D’Asseler
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
| | - Philippe Deron
- Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (L.B.); (P.D.); (W.H.)
- Department of Head and Neck Surgery, Ghent University Hospital, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
| | - Vincent Keereman
- Department of Electronics and Information Systems, Ghent University, 9000 Ghent, Belgium;
- XEOS Medical NV, 9000 Ghent, Belgium
| | - Koen Van de Vijver
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
- Department of Pathology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Christian Vanhove
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
- Department of Electronics and Information Systems, Ghent University, 9000 Ghent, Belgium;
- INFINITY Lab, Ghent University, 9000 Ghent, Belgium
| | - Wouter Huvenne
- Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (L.B.); (P.D.); (W.H.)
- Department of Head and Neck Surgery, Ghent University Hospital, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium; (V.S.); (D.C.); (Y.D.); (K.V.d.V.); (C.V.)
| |
Collapse
|
32
|
Hernandez Vargas S, Lin C, Tran Cao HS, Ikoma N, AghaAmiri S, Ghosh SC, Uselmann AJ, Azhdarinia A. Receptor-Targeted Fluorescence-Guided Surgery With Low Molecular Weight Agents. Front Oncol 2021; 11:674083. [PMID: 34277418 PMCID: PMC8279813 DOI: 10.3389/fonc.2021.674083] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer surgery remains the primary treatment option for most solid tumors and can be curative if all malignant cells are removed. Surgeons have historically relied on visual and tactile cues to maximize tumor resection, but clinical data suggest that relapse occurs partially due to incomplete cancer removal. As a result, the introduction of technologies that enhance the ability to visualize tumors in the operating room represents a pressing need. Such technologies have the potential to revolutionize the surgical standard-of-care by enabling real-time detection of surgical margins, subclinical residual disease, lymph node metastases and synchronous/metachronous tumors. Fluorescence-guided surgery (FGS) in the near-infrared (NIRF) spectrum has shown tremendous promise as an intraoperative imaging modality. An increasing number of clinical studies have demonstrated that tumor-selective FGS agents can improve the predictive value of fluorescence over non-targeted dyes. Whereas NIRF-labeled macromolecules (i.e., antibodies) spearheaded the widespread clinical translation of tumor-selective FGS drugs, peptides and small-molecules are emerging as valuable alternatives. Here, we first review the state-of-the-art of promising low molecular weight agents that are in clinical development for FGS; we then discuss the significance, application and constraints of emerging tumor-selective FGS technologies.
Collapse
Affiliation(s)
- Servando Hernandez Vargas
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Therapeutics & Pharmacology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | | | - Hop S Tran Cao
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naruhiko Ikoma
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Solmaz AghaAmiri
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sukhen C Ghosh
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | - Ali Azhdarinia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Therapeutics & Pharmacology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
33
|
Stroet MCM, Dijkstra BM, Dulfer SE, Kruijff S, den Dunnen WFA, Kruyt FAE, Groen RJM, Seimbille Y, Panth KM, Mezzanotte L, Lowik CWGM, de Jong M. Necrosis binding of Ac-Lys 0(IRDye800CW)-Tyr 3-octreotate: a consequence from cyanine-labeling of small molecules. EJNMMI Res 2021; 11:47. [PMID: 33970376 PMCID: PMC8110618 DOI: 10.1186/s13550-021-00789-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022] Open
Abstract
Background There is a growing body of nuclear contrast agents that are repurposed for fluorescence-guided surgery. New contrast agents are obtained by substituting the radioactive tag with, or adding a fluorescent cyanine to the molecular structure of antibodies or peptides. This enables intra-operative fluorescent detection of cancerous tissue, leading to more complete tumor resection. However, these fluorescent cyanines can have a remarkable influence on pharmacokinetics and tumor uptake, especially when labeled to smaller targeting vectors such as peptides. Here we demonstrate the effect of cyanine-mediated dead cell-binding of Ac-Lys0(IRDye800CW)-Tyr3-octreotate (800CW-TATE) and how this can be used as an advantage for fluorescence-guided surgery. Results Binding of 800CW-TATE could be blocked with DOTA0-Tyr3-octreotate (DOTA-TATE) on cultured SSTR2-positive U2OS cells and was absent in SSTR2 negative U2OS cells. However, strong binding was observed to dead cells, which could not be blocked with DOTA-TATE and was also present in dead SSTR2 negative cells. No SSTR2-mediated binding was observed in frozen tumor sections, possibly due to disruption of the cells in the process of sectioning the tissue before exposure to the contrast agent. DOTA-TATE blocking resulted in an incomplete reduction of 61.5 ± 5.8% fluorescence uptake by NCI-H69-tumors in mice. Near-infrared imaging and dead cell staining on paraffin sections from resected tumors revealed that fluorescence uptake persisted in necrotic regions upon blocking with DOTA-TATE. Conclusion This study shows that labeling peptides with cyanines can result in dead cell binding. This does not hamper the ultimate purpose of fluorescence-guided surgery, as necrotic tissue appears in most solid tumors. Hence, the necrosis binding can increase the overall tumor uptake. Moreover, necrotic tissue should be removed as much as possible: it cannot be salvaged, causes inflammation, and is tumorigenic. However, when performing binding experiments to cells with disrupted membrane integrity, which is routinely done with nuclear probes, this dead cell-binding can resemble non-specific binding. This study will benefit the development of fluorescent contrast agents. Supplementary information The online version contains supplementary material available at 10.1186/s13550-021-00789-4.
Collapse
Affiliation(s)
- Marcus C M Stroet
- Department of Radiology and Nuclear Medicine/Molecular Genetics, Erasmus Medical Centre, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands. .,Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.
| | - Bianca M Dijkstra
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sebastiaan E Dulfer
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Schelto Kruijff
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frank A E Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rob J M Groen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine/Molecular Genetics, Erasmus Medical Centre, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Kranthi M Panth
- Department of Radiology and Nuclear Medicine/Molecular Genetics, Erasmus Medical Centre, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands.,Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine/Molecular Genetics, Erasmus Medical Centre, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands.,Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Clemens W G M Lowik
- Department of Radiology and Nuclear Medicine/Molecular Genetics, Erasmus Medical Centre, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands.,CHUV Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine/Molecular Genetics, Erasmus Medical Centre, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| |
Collapse
|
34
|
Wang R, Alvarez DA, Crouch BT, Pilani A, Lam C, Zhu C, Hughes P, Katz D, Haystead T, Ramanujam N. Understanding the sources of errors in ex vivo Hsp90 molecular imaging for rapid-on-site breast cancer diagnosis. BIOMEDICAL OPTICS EXPRESS 2021; 12:2299-2311. [PMID: 33996230 PMCID: PMC8086448 DOI: 10.1364/boe.418818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 05/12/2023]
Abstract
Overexpression of heat shock protein 90 (Hsp90) on the surface of breast cancer cells makes it an attractive molecular biomarker for breast cancer diagnosis. Before a ubiquitous diagnostic method can be established, an understanding of the systematic errors in Hsp90-based imaging is essential. In this study, we investigated three factors that may influence the sensitivity of ex vivo Hsp90 molecular imaging: time-dependent tissue viability, nonspecific diffusion of an Hsp90 specific probe (HS-27), and contact-based imaging. These three factors will be important considerations when designing any diagnostic imaging strategy based on fluorescence imaging of a molecular target on tissue samples.
Collapse
Affiliation(s)
- Roujia Wang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Daniel A. Alvarez
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Brian T. Crouch
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Aditi Pilani
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Christopher Lam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Caigang Zhu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
- Currently at Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky, 40506, USA
| | - Philip Hughes
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, North Carolina 27708, USA
| | - David Katz
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Timothy Haystead
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, North Carolina 27708, USA
| | - Nirmala Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
35
|
Mao H, Long Y, Sheng X, Zhou X, Zhou B. Progress in application of near-infrared fluorescence imaging in the diagnosis and treatment of oral cancer. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:316-321. [PMID: 33927080 PMCID: PMC10929926 DOI: 10.11817/j.issn.1672-7347.2021.190762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Indexed: 11/03/2022]
Abstract
The preliminary screening of oral cancer mostly depends on the experience of clinicians, The surgical margin of tumor is mostly based on physical examination and preoperative imaging examination. It lacks real-time and objective intraoperative evaluation methods. Indocyanine green (ICG), as a safe and pollution-free organic fluorescent pigments, combined with near-infrared fluorescence imaging can be applied in the screening of early oral cancer, the determination of tumor resection margins, sentinel lymph node biopsy, cervical lymph node dissection, targeted chemotherapy, and other aspects. Near-infrared fluorescence imaging may become a key link in the early diagnosis and accurate treatment for oral cancer in the future.
Collapse
Affiliation(s)
- Huangxing Mao
- Breast Oncoplastic Surgery Department/ Head and Neck Surgery Department 3, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006.
| | - Ying Long
- Center for Translational Medicine, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Xiaowu Sheng
- Center for Translational Medicine, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| | - Xiao Zhou
- Breast Oncoplastic Surgery Department/ Head and Neck Surgery Department 3, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006
| | - Bo Zhou
- Breast Oncoplastic Surgery Department/ Head and Neck Surgery Department 3, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006.
| |
Collapse
|
36
|
Molecular imaging of a fluorescent antibody against epidermal growth factor receptor detects high-grade glioma. Sci Rep 2021; 11:5710. [PMID: 33707521 PMCID: PMC7952570 DOI: 10.1038/s41598-021-84831-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/16/2021] [Indexed: 01/31/2023] Open
Abstract
The prognosis for high-grade glioma (HGG) remains dismal and the extent of resection correlates with overall survival and progression free disease. Epidermal growth factor receptor (EGFR) is a biomarker heterogeneously expressed in HGG. We assessed the feasibility of detecting HGG using near-infrared fluorescent antibody targeting EGFR. Mice bearing orthotopic HGG xenografts with modest EGFR expression were imaged in vivo after systemic panitumumab-IRDye800 injection to assess its tumor-specific uptake macroscopically over 14 days, and microscopically ex vivo. EGFR immunohistochemical staining of 59 tumor specimens from 35 HGG patients was scored by pathologists and expression levels were compared to that of mouse xenografts. Intratumoral distribution of panitumumab-IRDye800 correlated with near-infrared fluorescence and EGFR expression. Fluorescence distinguished tumor cells with 90% specificity and 82.5% sensitivity. Target-to-background ratios peaked at 14 h post panitumumab-IRDye800 infusion, reaching 19.5 in vivo and 7.6 ex vivo, respectively. Equivalent or higher EGFR protein expression compared to the mouse xenografts was present in 77.1% HGG patients. Age, combined with IDH-wildtype cerebral tumor, was predictive of greater EGFR protein expression in human tumors. Tumor specific uptake of panitumumab-IRDye800 provided remarkable contrast and a flexible imaging window for fluorescence-guided identification of HGGs despite modest EGFR expression.
Collapse
|
37
|
Pogue BW, Rosenthal EL. Review of successful pathways for regulatory approvals in open-field fluorescence-guided surgery. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-210023VR. [PMID: 33715318 PMCID: PMC7955139 DOI: 10.1117/1.jbo.26.3.030901] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/23/2021] [Indexed: 06/12/2023]
Abstract
SIGNIFICANCE The modern use of fluorescence in surgery came iteratively through new devices and pre-existing imaging agents, with indications that were paved via regulatory approvals and device clearances. These events led to a growing set of surgery subspecialty uses. AIM This article outlines the key milestones that initiated commercially marketed systems and agents by highlighting temporal sequences and strategic decisions between them, with the goal of helping to inform future successes. APPROACH A review of successful regulatory approvals and the sequences between them was completed for companies that achieved US Food and Drug Administration (FDA) premarket approval or new drug approvals (NDAs) or device clearances in the fields of fluorescent imaging agents, open surgery imaging devices, and their approved medical indications. RESULTS Angiography agents, indocyanine green and fluorescein, were approved for human use as absorbing dyes, and this use in retinal imaging was the precursor to lateral translation into tissue perfusion imaging in the last two decades with a growing number of devices. Many FDA cleared devices for open fluorescence-guided surgery used the predicate created by the SPY SP2000 system. This first system was 510(k) cleared for angiography imaging with a unique split predicate from x-ray imaging of vasculature and retinal fluorescence angiography. Since that time, the lateral spread of open surgery devices being cleared for new indications has been occurring with a growth of adoption in surgical subspecialties. Growth into new surgical subspecialties has been achieved by leveraging different NDAs and clearances between indications, such that medical uses have broadened over time. CONCLUSIONS Key decisions made by developers to advance specific device clearances and NDAs have been based upon existing optical fluorescent agents. The historical lessons and regulatory trends in newer indications and contrast agents can help the field evolve via successful investment in new systems and applications.
Collapse
Affiliation(s)
- Brian W. Pogue
- Thayer School of Engineering at Dartmouth, Center for Imaging Medicine, Hanover, New Hampshire, United States
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States
| | - Eben L. Rosenthal
- Stanford University School of Medicine, Palo Alto, California, United States
| |
Collapse
|
38
|
Unique Benefits of Tumor-Specific Nanobodies for Fluorescence Guided Surgery. Biomolecules 2021; 11:biom11020311. [PMID: 33670740 PMCID: PMC7921980 DOI: 10.3390/biom11020311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor-specific fluorescence labeling is promising for real-time visualization of solid malignancies during surgery. There are a number of technologies to confer tumor-specific fluorescence. Antibodies have traditionally been used due to their versatility in modifications; however, their large size hampers efficient fluorophore delivery. Nanobodies are a novel class of molecules, derived from camelid heavy-chain only antibodies, that have shown promise for tumor-specific fluorescence labeling. Nanobodies are ten times smaller than standard antibodies, while maintaining antigen-binding capacity and have advantageous features, including rapidity of tumor labeling, that are reviewed in the present report. The present report reviews special considerations needed in developing nanobody probes, the status of current literature on the use of nanobody probes in fluorescence guided surgery, and potential challenges to be addressed for clinical translation.
Collapse
|
39
|
Bhandari C, Guirguis M, Savan NA, Shrivastava N, Oliveira S, Hasan T, Obaid G. What NIR photodynamic activation offers molecular targeted nanomedicines: Perspectives into the conundrum of tumor specificity and selectivity. NANO TODAY 2021; 36:101052. [PMID: 33552231 PMCID: PMC7864390 DOI: 10.1016/j.nantod.2020.101052] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Near infrared (NIR) photodynamic activation is playing increasingly critical roles in cutting-edge anti-cancer nanomedicines, which include spatiotemporal control over induction of therapy, photodynamic priming, and phototriggered immunotherapy. Molecular targeted photonanomedicines (mt-PNMs) are tumor-specific nanoscale drug delivery systems, which capitalize on the unparalleled spatio-temporal precision of NIR photodynamic activation to augment the accuracy of tumor tissue treatment. mt-PNMs are emerging as a paradigm approach for the targeted treatment of solid tumors, yet remain highly complex and multifaceted. While ligand targeted nanomedicines in general suffer from interdependent challenges in biophysics, surface chemistry and nanotechnology, mt-PNMs provide distinct opportunities to synergistically potentiate the effects of ligand targeting. This review provides what we believe to be a much-need demarcation between the processes involved in tumor specificity (biomolecular recognition events) and tumor selectivity (preferential tumor accumulation) of ligand targeted nanomedicines, such as mt-PNMs, and elaborate on what NIR photodynamic activation has to offer. We discuss the interplay between both tumor specificity and tumor selectivity and the degree to which both may play central roles in cutting-edge NIR photoactivable nanotechnologies. A special emphasis is made on NIR photoactivable biomimetic nanotechnologies that capitalize on both specificity and selectivity phenomena to augment the safety and efficacy of photodynamic anti-tumor regimens.
Collapse
Affiliation(s)
- Chanda Bhandari
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| | - Mina Guirguis
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| | - N. Anna Savan
- Michigan State University, East Lansing, Michigan, 48824, U.S
| | - Navadeep Shrivastava
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| | - Sabrina Oliveira
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands
- Pharmaceutics, Department of Pharmaceutical Sciences, Science Faculty, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, U.S
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, U.S
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, U.S
| |
Collapse
|
40
|
Achterberg FB, Deken MM, Meijer RPJ, Mieog JSD, Burggraaf J, van de Velde CJH, Swijnenburg RJ, Vahrmeijer AL. Clinical translation and implementation of optical imaging agents for precision image-guided cancer surgery. Eur J Nucl Med Mol Imaging 2021; 48:332-339. [PMID: 32783112 PMCID: PMC7835299 DOI: 10.1007/s00259-020-04970-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The field of tumor-specific fluorescence-guided surgery has seen a significant increase in the development of novel tumor-targeted imaging agents. Studying patient benefit using intraoperative fluorescence-guided imaging for cancer surgery is the final step needed for implementation in standard treatment protocols. Translation into phase III clinical trials can be challenging and time consuming. Recent studies have helped to identify certain waypoints in this transition phase between studying imaging agent efficacy (phase I-II) and proving patient benefit (phase III). TRIAL INITIATION Performing these trials outside centers of expertise, thus involving motivated clinicians, training them, and providing feedback on data quality, increases the translatability of imaging agents and the surgical technique. Furthermore, timely formation of a trial team which oversees the translational process is vital. They are responsible for establishing an imaging framework (camera system, imaging protocol, surgical workflow) and clinical framework (disease stage, procedure type, clinical research question) in which the trial is executed. Providing participating clinicians with well-defined protocols with the aim to answer clinically relevant research questions within the context of care is the pinnacle in gathering reliable trial data. OUTLOOK If all these aspects are taken into consideration, tumor-specific fluorescence-guided surgery is expected be of significant value when integrated into the diagnostic work-up, surgical procedure, and follow-up of cancer patients. It is only by involving and collaborating with all stakeholders involved in this process that successful clinical translation can occur. AIM Here, we discuss the challenges faced during this important translational phase and present potential solutions to enable final clinical translation and implementation of imaging agents for image-guided cancer surgery.
Collapse
Affiliation(s)
- F B Achterberg
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - M M Deken
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - R P J Meijer
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J S D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J Burggraaf
- Centre for Human Drug Research (CHDR), Leiden, The Netherlands
| | - C J H van de Velde
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - R J Swijnenburg
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
41
|
Sier VQ, de Vries MR, van der Vorst JR, Vahrmeijer AL, van Kooten C, Cruz LJ, de Geus-Oei LF, Ferreira V, Sier CFM, Alves F, Muthana M. Cell-Based Tracers as Trojan Horses for Image-Guided Surgery. Int J Mol Sci 2021; 22:E755. [PMID: 33451116 PMCID: PMC7828607 DOI: 10.3390/ijms22020755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022] Open
Abstract
Surgeons rely almost completely on their own vision and palpation to recognize affected tissues during surgery. Consequently, they are often unable to distinguish between different cells and tissue types. This makes accurate and complete resection cumbersome. Targeted image-guided surgery (IGS) provides a solution by enabling real-time tissue recognition. Most current targeting agents (tracers) consist of antibodies or peptides equipped with a radiolabel for Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT), magnetic resonance imaging (MRI) labels, or a near-infrared fluorescent (NIRF) dye. These tracers are preoperatively administered to patients, home in on targeted cells or tissues, and are visualized in the operating room via dedicated imaging systems. Instead of using these 'passive' tracers, there are other, more 'active' approaches of probe delivery conceivable by using living cells (macrophages/monocytes, neutrophils, T cells, mesenchymal stromal cells), cell(-derived) fragments (platelets, extracellular vesicles (exosomes)), and microorganisms (bacteria, viruses) or, alternatively, 'humanized' nanoparticles. Compared with current tracers, these active contrast agents might be more efficient for the specific targeting of tumors or other pathological tissues (e.g., atherosclerotic plaques). This review provides an overview of the arsenal of possibilities applicable for the concept of cell-based tracers for IGS.
Collapse
Affiliation(s)
- Vincent Q. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Margreet R. de Vries
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Joost R. van der Vorst
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Cornelis van Kooten
- Department of Nephrology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Luis J. Cruz
- Department of Radiology, Translational Nanomaterials and Imaging Group, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
| | - Valerie Ferreira
- Department of Research and Development, UniQure, 1100 DA Amsterdam, The Netherlands;
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
- Percuros B.V. Leiden, 2333 CL Leiden, The Netherlands
| | - Frauke Alves
- Translational Molecular Imaging, Clinic of Hematology and Medical Oncology, Institute of Diagnostic and Interventional Radiology, University Medicine Center Göttingen and Max-Planck-Institute for Experimental Medicine, 37075 Göttingen, Germany;
| | - Munitta Muthana
- Department of Infection and Immunity, University of Sheffield, Sheffield S10 2RX, UK;
| |
Collapse
|
42
|
Gorpas D, Ntziachristos V, Tian J. Principles and Practice of Intraoperative Fluorescence Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00009-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
43
|
Curtin N, Wu D, Cahill R, Sarkar A, Aonghusa PM, Zhuk S, Barberio M, Al-Taher M, Marescaux J, Diana M, O'Shea DF. Dual Color Imaging from a Single BF 2-Azadipyrromethene Fluorophore Demonstrated in vivo for Lymph Node Identification. Int J Med Sci 2021; 18:1541-1553. [PMID: 33746570 PMCID: PMC7976573 DOI: 10.7150/ijms.52816] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
Dual emissions at ~700 and 800 nm have been achieved from a single NIR-AZA fluorophore 1 by establishing parameters in which it can exist in either its isolated molecular or aggregated states. Dual near infrared (NIR) fluorescence color lymph node (LN) mapping with 1 was achieved in a large-animal porcine model, with injection site, channels and nodes all detectable at both 700 and 800 nm using a preclinical open camera system. The fluorophore was also compatible with imaging using two clinical instruments for fluorescence guided surgery. Methods: An NIR-AZA fluorophore with hydrophilic and phobic features was synthesised in a straightforward manner and its aggregation properties characterised spectroscopically and by TEM imaging. Toxicity was assessed in a rodent model and dual color fluorescence imaging evaluated by lymph node mapping in a large animal porcine models and in ex-vivo human tissue specimen. Results: Dual color fluorescence imaging has been achieved in the highly complex biomedical scenario of lymph node mapping. Emissions at 700 and 800 nm can be achieved from a single fluorophore by establishing molecular and aggregate forms. Fluorophore was compatible with clinical systems for fluorescence guided surgery and no toxicity was observed in high dosage testing. Conclusion: A new, biomedical compatible form of NIR-dual emission wavelength imaging has been established using a readily accessible fluorophore with significant scope for clinical translation.
Collapse
Affiliation(s)
- Niamh Curtin
- Department of Chemistry, Royal College of Surgeons in Ireland, RCSI, 123 St Stephen's Green, Dublin 2, Ireland
| | - Dan Wu
- Department of Chemistry, Royal College of Surgeons in Ireland, RCSI, 123 St Stephen's Green, Dublin 2, Ireland
| | - Ronan Cahill
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, Ireland; Department of Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Anwesha Sarkar
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, Ireland; Department of Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Pól Mac Aonghusa
- IBM Research - Ireland, Damastown Industrial Estate, Mulhuddart, Dublin 15, Ireland
| | - Sergiy Zhuk
- IBM Research - Ireland, Damastown Industrial Estate, Mulhuddart, Dublin 15, Ireland
| | - Manuel Barberio
- IHU-Strasbourg, Institute of Hybrid Image-Guided Surgery, Strasbourg, France
| | - Mahdi Al-Taher
- IHU-Strasbourg, Institute of Hybrid Image-Guided Surgery, Strasbourg, France
| | - Jacques Marescaux
- IRCAD, Research Institute against Cancer of the Digestive System, Strasbourg, France
| | - Michele Diana
- IHU-Strasbourg, Institute of Hybrid Image-Guided Surgery, Strasbourg, France.,IRCAD, Research Institute against Cancer of the Digestive System, Strasbourg, France.,ICube Lab, Photonics Instrumentation for Health, Strasbourg, France
| | - Donal F O'Shea
- Department of Chemistry, Royal College of Surgeons in Ireland, RCSI, 123 St Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
44
|
Hernandez Vargas S, Lin C, Voss J, Ghosh SC, Halperin DM, AghaAmiri S, Cao HST, Ikoma N, Uselmann AJ, Azhdarinia A. Development of a drug-device combination for fluorescence-guided surgery in neuroendocrine tumors. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200129R. [PMID: 33300316 PMCID: PMC7725236 DOI: 10.1117/1.jbo.25.12.126002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/06/2020] [Indexed: 05/13/2023]
Abstract
SIGNIFICANCE The use of cancer-targeted contrast agents in fluorescence-guided surgery (FGS) has the potential to improve intraoperative visualization of tumors and surgical margins. However, evaluation of their translational potential is challenging. AIM We examined the utility of a somatostatin receptor subtype-2 (SSTR2)-targeted fluorescent agent in combination with a benchtop near-infrared fluorescence (NIRF) imaging system to visualize mouse xenografts under conditions that simulate the clinical FGS workflow for open surgical procedures. APPROACH The dual-labeled somatostatin analog, Ga67-MMC(IR800)-TOC, was injected into mice (n = 24) implanted with SSTR2-expressing tumors and imaged with the customized OnLume NIRF imaging system (Madison, Wisconsin). In vivo and ex vivo imaging were performed under ambient light. The optimal dose (0.2, 0.5, and 2 nmol) and imaging time point (3, 24, 48, and 72 h) were determined using contrast-to-noise ratio (CNR) as the image quality parameter. Video captures of tumor resections were obtained to provide an FGS readout that is representative of clinical utility. Finally, a log-transformed linear regression model was fitted to assess congruence between fluorescence readouts and the underlying drug distribution. RESULTS The drug-device combination provided high in vivo and ex vivo contrast (CNRs > 3, except lung at 3 h) at all time points with the optimal dose of 2 nmol. The optimal imaging time point was 24-h post-injection, where CNRs > 6.5 were achieved in tissues of interest (i.e., pancreas, small intestine, stomach, and lung). Intraoperative FGS showed excellent utility for examination of the tumor cavity pre- and post-resection. The relationship between fluorescence readouts and gamma counts was linear and strongly correlated (n = 334, R2 = 0.71; r = 0.84; P < 0.0001). CONCLUSION The innovative OnLume NIRF imaging system enhanced the evaluation of Ga67-MMC(IR800)-TOC in tumor models. These components comprise a promising drug-device combination for FGS in patients with SSTR2-expressing tumors.
Collapse
Affiliation(s)
- Servando Hernandez Vargas
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| | | | - Julie Voss
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| | - Sukhen C. Ghosh
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| | - Daniel M. Halperin
- The University of Texas MD Anderson Cancer Center, Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, Houston, Texas, United States
| | - Solmaz AghaAmiri
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| | - Hop S. Tran Cao
- The University of Texas MD Anderson Cancer Center, Department of Surgical Oncology, Division of Surgery, Houston, Texas, United States
| | - Naruhiko Ikoma
- The University of Texas MD Anderson Cancer Center, Department of Surgical Oncology, Division of Surgery, Houston, Texas, United States
| | | | - Ali Azhdarinia
- The University of Texas Health Science Center at Houston, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, Houston, Texas, United States
| |
Collapse
|
45
|
Ashoka AH, Kong SH, Seeliger B, Andreiuk B, Soares RV, Barberio M, Diana M, Klymchenko AS. Near-infrared fluorescent coatings of medical devices for image-guided surgery. Biomaterials 2020; 261:120306. [DOI: 10.1016/j.biomaterials.2020.120306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
|
46
|
Sadeghipour N, Rangnekar A, Folaron MR, Strawbridge RR, Samkoe KS, Davis SC, Tichauer KM. Prediction of optimal contrast times post-imaging agent administration to inform personalized fluorescence-guided surgery. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200182RR. [PMID: 33200596 PMCID: PMC7667427 DOI: 10.1117/1.jbo.25.11.116005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/30/2020] [Indexed: 05/08/2023]
Abstract
SIGNIFICANCE Fluorescence guidance in cancer surgery (FGS) using molecular-targeted contrast agents is accelerating, yet the influence of individual patients' physiology on the optimal time to perform surgery post-agent-injection is not fully understood. AIM Develop a mathematical framework and analytical expressions to estimate patient-specific time-to-maximum contrast after imaging agent administration for single- and paired-agent (coadministration of targeted and control agents) protocols. APPROACH The framework was validated in mouse subcutaneous xenograft studies for three classes of imaging agents: peptide, antibody mimetic, and antibody. Analytical expressions estimating time-to-maximum-tumor-discrimination potential were evaluated over a range of parameters using the validated framework for human cancer parameters. RESULTS Correlations were observed between simulations and matched experiments and metrics of tumor discrimination potential (p < 0.05). Based on human cancer physiology, times-to-maximum contrast for peptide and antibody mimetic agents were <200 min, >15 h for antibodies, on average. The analytical estimates of time-to-maximum tumor discrimination performance exhibited errors of <10 % on average, whereas patient-to-patient variance is expected to be greater than 100%. CONCLUSION We demonstrated that analytical estimates of time-to-maximum contrast in FGS carried out patient-to-patient can outperform the population average time-to-maximum contrast used currently in clinical trials. Such estimates can be made with preoperative DCE-MRI (or similar) and knowledge of the targeted agent's binding affinity.
Collapse
Affiliation(s)
- Negar Sadeghipour
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
- Stanford University School of Medicine, Molecular Imaging Program at Stanford, Palo Alto, California, United States
- Stanford University School of Medicine, Canary Center at Stanford for Cancer Early Detection, Palo Alto, California, United States
| | - Aakanksha Rangnekar
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Margaret R. Folaron
- Dartmouth College, Thayer School for Engineering, Hanover, New Hampshire, United States
| | | | - Kimberley S. Samkoe
- Dartmouth College, Thayer School for Engineering, Hanover, New Hampshire, United States
| | - Scott C. Davis
- Dartmouth College, Thayer School for Engineering, Hanover, New Hampshire, United States
| | - Kenneth M. Tichauer
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| |
Collapse
|
47
|
A Photocleavable Contrast Agent for Light-Responsive MRI. Pharmaceuticals (Basel) 2020; 13:ph13100296. [PMID: 33050049 PMCID: PMC7599822 DOI: 10.3390/ph13100296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/18/2020] [Accepted: 10/03/2020] [Indexed: 11/16/2022] Open
Abstract
Thanks to its innocuousness and high spatiotemporal resolution, light is used in several established and emerging applications in biomedicine. Among them is the modulation of magnetic resonance imaging (MRI) contrast agents’ relaxivity with the aim to increase the sensitivity, selectivity and amount of functional information obtained from this outstanding whole-body medical imaging technique. This approach requires the development of molecular contrast agents that show high relaxivity and strongly pronounced photo-responsiveness. To this end, we report here the design and synthesis of a light-activated MRI contrast agent, together with its evaluation using UV–vis spectroscopy, Fast Field Cycling (FFC) relaxometry and relaxometric measurements on clinical MRI scanners. The high relaxivity of the reported agent changes substantially upon irradiation with light, showing a 17% decrease in relaxivity at 0.23T upon irradiation with λ = 400 nm (violet) light for 60 min. On clinical MRI scanners (1.5T and 3.0T), irradiation leads to a decrease in relaxivity of 9% and 19% after 3 and 60 min, respectively. The molecular design presents an important blueprint for the development of light-activatable MRI contrast agents.
Collapse
|
48
|
Wojtynek NE, Mohs AM. Image-guided tumor surgery: The emerging role of nanotechnology. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1624. [PMID: 32162485 PMCID: PMC9469762 DOI: 10.1002/wnan.1624] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/15/2022]
Abstract
Surgical resection is a mainstay treatment for solid tumors. Yet, methods to distinguish malignant from healthy tissue are primarily limited to tactile and visual cues as well as the surgeon's experience. As a result, there is a possibility that a positive surgical margin (PSM) or the presence of residual tumor left behind after resection may occur. It is well-documented that PSMs can negatively impact treatment outcomes and survival, as well as pose an economic burden. Therefore, surgical tumor imaging techniques have emerged as a promising method to decrease PSM rates. Nanoparticles (NPs) have unique characteristics to serve as optical contrast agents during image-guided surgery (IGS). Recently, there has been tremendous growth in the volume and types of NPs used for IGS, including clinical trials. Herein, we describe the most recent contributions of nanotechnology for surgical tumor identification. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Implantable Materials and Surgical Technologies > Nanoscale Tools and Techniques in Surgery Diagnostic Tools > in vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Nicholas E. Wojtynek
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Aaron M. Mohs
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
49
|
Beyer T, Bidaut L, Dickson J, Kachelriess M, Kiessling F, Leitgeb R, Ma J, Shiyam Sundar LK, Theek B, Mawlawi O. What scans we will read: imaging instrumentation trends in clinical oncology. Cancer Imaging 2020; 20:38. [PMID: 32517801 PMCID: PMC7285725 DOI: 10.1186/s40644-020-00312-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
Oncological diseases account for a significant portion of the burden on public healthcare systems with associated costs driven primarily by complex and long-lasting therapies. Through the visualization of patient-specific morphology and functional-molecular pathways, cancerous tissue can be detected and characterized non-invasively, so as to provide referring oncologists with essential information to support therapy management decisions. Following the onset of stand-alone anatomical and functional imaging, we witness a push towards integrating molecular image information through various methods, including anato-metabolic imaging (e.g., PET/CT), advanced MRI, optical or ultrasound imaging.This perspective paper highlights a number of key technological and methodological advances in imaging instrumentation related to anatomical, functional, molecular medicine and hybrid imaging, that is understood as the hardware-based combination of complementary anatomical and molecular imaging. These include novel detector technologies for ionizing radiation used in CT and nuclear medicine imaging, and novel system developments in MRI and optical as well as opto-acoustic imaging. We will also highlight new data processing methods for improved non-invasive tissue characterization. Following a general introduction to the role of imaging in oncology patient management we introduce imaging methods with well-defined clinical applications and potential for clinical translation. For each modality, we report first on the status quo and, then point to perceived technological and methodological advances in a subsequent status go section. Considering the breadth and dynamics of these developments, this perspective ends with a critical reflection on where the authors, with the majority of them being imaging experts with a background in physics and engineering, believe imaging methods will be in a few years from now.Overall, methodological and technological medical imaging advances are geared towards increased image contrast, the derivation of reproducible quantitative parameters, an increase in volume sensitivity and a reduction in overall examination time. To ensure full translation to the clinic, this progress in technologies and instrumentation is complemented by advances in relevant acquisition and image-processing protocols and improved data analysis. To this end, we should accept diagnostic images as "data", and - through the wider adoption of advanced analysis, including machine learning approaches and a "big data" concept - move to the next stage of non-invasive tumour phenotyping. The scans we will be reading in 10 years from now will likely be composed of highly diverse multi-dimensional data from multiple sources, which mandate the use of advanced and interactive visualization and analysis platforms powered by Artificial Intelligence (AI) for real-time data handling by cross-specialty clinical experts with a domain knowledge that will need to go beyond that of plain imaging.
Collapse
Affiliation(s)
- Thomas Beyer
- QIMP Team, Centre for Medical Physics and Biomedical Engineering, Medical University Vienna, Währinger Gürtel 18-20/4L, 1090, Vienna, Austria.
| | - Luc Bidaut
- College of Science, University of Lincoln, Lincoln, UK
| | - John Dickson
- Institute of Nuclear Medicine, University College London Hospital, London, UK
| | - Marc Kachelriess
- Division of X-ray imaging and CT, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, DE, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074, Aachen, DE, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Am Fallturm 1, 28359, Bremen, DE, Germany
| | - Rainer Leitgeb
- Centre for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, AT, Austria
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lalith Kumar Shiyam Sundar
- QIMP Team, Centre for Medical Physics and Biomedical Engineering, Medical University Vienna, Währinger Gürtel 18-20/4L, 1090, Vienna, Austria
| | - Benjamin Theek
- Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstrasse 20, 52074, Aachen, DE, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Am Fallturm 1, 28359, Bremen, DE, Germany
| | - Osama Mawlawi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
50
|
Fan D, Wang K, Gao H, Luo Q, Wang X, Li X, Tong W, Zhang X, Luo C, Yang G, Ai L, Shi J. A 64 Cu-porphyrin-based dual-modal molecular probe with integrin α v β 3 targeting function for tumour imaging. J Labelled Comp Radiopharm 2020; 63:212-221. [PMID: 32083750 DOI: 10.1002/jlcr.3833] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 02/01/2023]
Abstract
Pyropheophorbide-a (Pyro) is a promising multifunctional molecule for multimodal tumour imaging and photodynamic therapy, but its clinical applications are seriously restricted by the limited tumour accumulation capability. Here, we designed and synthesized a small-molecule probe that achieved specific dual-modal tumour imaging based on Pyro. Briefly, a novel molecule combining Pyro, an RGD dimer peptide (3PRGD2 ) and 64 Cu, was designed and synthesized, and the obtained molecule, 64 Cu-Pyro-3PRGD2 , exhibited high tumour specificity in both positron emission tomography and optical imaging in vivo. c (RGDfk) peptide blocking significantly reduced the efficacy of the probe, which confirmed the integrin αV β3 targeting of this molecular probe. 64 Cu-Pyro-3PRGD2 had very low accumulation in normal organs and could be rapidly cleared through kidney metabolism, which prevented the potential damage to adjacent normal tissues. Overall, combining tumour targeting, dual-modal imaging, and biosafety, 64 Cu-Pyro-3PRGD2 has the potential for clinical use as a molecular imaging probe for tumour diagnosis.
Collapse
Affiliation(s)
- Di Fan
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kai Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hannan Gao
- Medical Isotopes Research Center, Peking University, Beijing, China
| | - Qi Luo
- Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Xin Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaotong Li
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wu Tong
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Medical Isotopes Research Center, Peking University, Beijing, China
| | - Chuangwei Luo
- Medical Isotopes Research Center, Peking University, Beijing, China
| | - Guangjie Yang
- Medical Isotopes Research Center, Peking University, Beijing, China
| | - Lin Ai
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiyun Shi
- Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing, China
| |
Collapse
|