1
|
Singh N, Xia W, Need E, McManus K, Huang J, Shi S, Goel S. Tumor agnostic ultrasmall nanoprobes for fluorescence-guided surgical resection in peritoneal metastasis. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06950-0. [PMID: 39446146 DOI: 10.1007/s00259-024-06950-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE Surgical excision of metastases is the only curative treatment strategy in peritoneal carcinomatosis management, and the completeness of tumor resection determines the success of the surgery. Tumor-specific fluorescence-guided probes can improve the outcomes of cytoreductive surgery and thereby prognosis. This study aimed to develop and evaluate the feasibility of fluorescently labeled ultrasmall porous silica nanoparticles (UPSN) for image-guided resection of peritoneally disseminated tumors of different origins. METHODS Ultrasmall fluorescent nanoprobes were synthesized and characterized for their physicochemical properties and stability. Tumor-specific uptake and biodistribution profiles were evaluated in syngeneic CT26 colorectal and KPC-689 pancreatic cancer murine models. The practicability of real-time optical UPSN-guided resection was examined in the CT26 colorectal cancer model using a surgical stereomicroscope. Quantitative measurements of tumor sensitivity and specificity were performed. Histopathological examination validated in vivo findings about tumor-specific accumulation and safety of ultrasmall fluorescent probes. RESULTS As-synthesized UPSNs were successfully surface modified with Cy5 or Cy3 dyes maintaining sub-15 nm size and near neutral charge which is beneficial for optimized in vivo pharmacokinetics. UPSN-Cy5 demonstrated high tumor-specific uptake and favorable biodistribution profiles in peritoneal metastasis models of CT26 and KPC tumors. Dye-conjugated UPSN enabled resection of microscopic lesions and achieved a higher tumor-to-background ratios in comparison to FDA-approved indocyanine green (ICG) dye in both models. Microscopic evaluation showed tumor localization and off-target safety profile of the UPSN-Cy5. CONCLUSION Ultrasmall fluorescent probes were effective in surgical resection of peritoneal metastases with high sensitivity and specificity, thus emerging as promising tumor agnostic agents for image-guided cancer surgery.
Collapse
Affiliation(s)
- Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Esther Need
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Kylee McManus
- College of Science and Honors College (Biology), University of Utah, Salt Lake City, UT, 84112, USA
| | - Jiemin Huang
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA.
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA.
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT, 84112, USA.
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
2
|
Xu M, Li P, Wei J, Yan P, Zhang Y, Guo X, Liu C, Yang X. Progress of fluorescence imaging in lymph node dissection surgery for prostate and bladder cancer. Front Oncol 2024; 14:1395284. [PMID: 39429471 PMCID: PMC11486700 DOI: 10.3389/fonc.2024.1395284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Fluorescence imaging is a relatively new imaging method used to visualize different tissue structures to help guide intraoperative operations, which has potential advantages with high sensitivity and contrast compared to conventional imaging. In this work, we review fluorescent contrast agents and devices used for lymphatic system imaging. Indocyanine green is the most widely utilized due to its high sensitivity, specificity, low background fluorescence, and safety profile. In prostate and bladder cancer lymph node dissection, the complex lymphatic drainage can result in missed metastatic nodes and extensive dissection increases the risk of complications like lymphocele, presenting a significant challenge for urologists. Fluorescence-guided sentinel lymph node dissection facilitates precise tumor staging. The combination of fluorescence and radiographic imaging improves the accuracy of lymph node staging. Multimodal imaging presents new potential for precisely identifying metastatic pelvic lymph nodes.
Collapse
Affiliation(s)
- Mingquan Xu
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, ;China
| | - Panpan Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Jinzheng Wei
- Department of Orthopedics, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Pengyu Yan
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Yunmeng Zhang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Xinyu Guo
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
| | - Chao Liu
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, ;China
| | - Xiaofeng Yang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, ;China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, ;China
| |
Collapse
|
3
|
Lee KH, Cox KE, Amirfakhri S, Jaiswal S, Liu S, Hosseini M, Lwin TM, Yazaki PJ, Hoffman RM, Bouvet M. Accurate Co-Localization of Luciferase Expression and Fluorescent Anti-CEA Antibody Targeting of Liver Metastases in an Orthotopic Mouse Model of Colon Cancer. Cancers (Basel) 2024; 16:3341. [PMID: 39409961 PMCID: PMC11475688 DOI: 10.3390/cancers16193341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND The present study aimed to validate the accuracy of a tumor-specific antibody to target liver metastases of colorectal cancer. METHODS A humanized anti-CEA antibody conjugated to a fluorescent dye (M5A-IR800) was tested for targeting human colorectal cancer liver metastases (CRLMs) expressing luciferase in an orthotopic mouse model. Orthotopic mouse models of CRLMs were established by implanting fragments of a luciferase-expressing human colorectal cancer cell line, LS174T, in the liver of nude mice. Mice received 50 µg M5A-IR800 72 h prior to imaging. To test co-localization, bioluminescence imaging was performed using D-luciferin, which was given via intraperitoneal injection just prior to imaging. RESULTS Tumors were able to be visualized non-invasively through the skin with the luciferase-luciferin signal. Intra-abdominal imaging showed accurate labeling of CRLMs with M5A-IR800, which co-localized with the luciferase-luciferin signal. CONCLUSIONS The present results validate the accuracy of a tumor-specific anti-CEA antibody in targeting liver metastases of colorectal cancer.
Collapse
Affiliation(s)
- Kyung-Ha Lee
- Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA; (K.-H.L.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
- Department of Colorectal Surgery, Chungnam National University Hospital, Daejeon 35015, Republic of Korea
| | - Kristin E. Cox
- Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA; (K.-H.L.); (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 92037, USA; (K.-H.L.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Sunidhi Jaiswal
- Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA; (K.-H.L.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Shanglei Liu
- Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA; (K.-H.L.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Mojgan Hosseini
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Thinzar M. Lwin
- Department of Surgical Oncology, City of Hope, Duarte, CA 91010, USA
| | - Paul J. Yazaki
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA; (K.-H.L.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
- AntiCancer Inc., San Diego, CA 92111, USA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA; (K.-H.L.); (R.M.H.)
- VA San Diego Healthcare System, La Jolla, CA 92161, USA
| |
Collapse
|
4
|
Pace J, Lee JJ, Srinivasarao M, Kallepu S, Low PS, Niedre M. In Vivo Labeling and Detection of Circulating Tumor Cells in Mice Using OTL38. Mol Imaging Biol 2024; 26:603-615. [PMID: 38594545 PMCID: PMC11281960 DOI: 10.1007/s11307-024-01914-0] [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: 12/12/2023] [Revised: 03/04/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE We recently developed an optical instrument to non-invasively detect fluorescently labeled circulating tumor cells (CTCs) in mice called 'Diffuse in vivo Flow Cytometry' (DiFC). OTL38 is a folate receptor (FR) targeted near-infrared (NIR) contrast agent that is FDA approved for use in fluorescence guided surgery of ovarian and lung cancer. In this work, we investigated the use OTL38 for in vivo labeling and detection of FR + CTCs with DiFC. PROCEDURES We tested OTL38 labeling of FR + cancer cell lines (IGROV-1 and L1210A) as well as FR- MM.1S cells in suspensions of Human Peripheral Blood Mononuclear cells (PBMCs) in vitro. We also tested OTL38 labeling and NIR-DIFC detection of FR + L1210A cells in blood circulation in nude mice in vivo. RESULTS 62% of IGROV-1 and 83% of L1210A were labeled above non-specific background levels in suspensions of PBMCs in vitro compared to only 2% of FR- MM.1S cells. L1210A cells could be labeled with OTL38 directly in circulation in vivo and externally detected using NIR-DiFC in mice with low false positive detection rates. CONCLUSIONS This work shows the feasibility of labeling CTCs in vivo with OTL38 and detection with DiFC. Although further refinement of the DiFC instrument and signal processing algorithms and testing with other animal models is needed, this work may eventually pave the way for human use of DiFC.
Collapse
Affiliation(s)
- Joshua Pace
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Jane J Lee
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | | | | | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, 047906, USA
| | - Mark Niedre
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Hadzima M, Faucher FF, Blažková K, Yim JJ, Guerra M, Chen S, Woods EC, Park KW, Šácha P, Šubr V, Kostka L, Etrych T, Majer P, Konvalinka J, Bogyo M. Polymer-Tethered Quenched Fluorescent Probes for Enhanced Imaging of Tumor-Associated Proteases. ACS Sens 2024; 9:3720-3729. [PMID: 38941307 PMCID: PMC11287742 DOI: 10.1021/acssensors.4c00912] [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: 04/18/2024] [Revised: 05/30/2024] [Accepted: 06/12/2024] [Indexed: 06/30/2024]
Abstract
Fluorescence-based contrast agents enable real-time detection of solid tumors and their neovasculature, making them ideal for use in image-guided surgery. Several agents have entered late-stage clinical trials or secured FDA approval, suggesting they are likely to become the standard of care in cancer surgeries. One of the key parameters to optimize in contrast agents is molecular size, which dictates much of the pharmacokinetic and pharmacodynamic properties of the agent. Here, we describe the development of a class of protease-activated quenched fluorescent probes in which a N-(2-hydroxypropyl)methacrylamide copolymer is used as the primary scaffold. This copolymer core provides a high degree of probe modularity to generate structures that cannot be achieved with small molecules and peptide probes. We used a previously validated cathepsin substrate and evaluated the effects of length and type of linker, as well as the positioning of the fluorophore/quencher pair on the polymer core. We found that the polymeric probes could be optimized to achieve increased overall signal and tumor-to-background ratios compared to the reference small molecule probe. Our results also revealed multiple structure-activity relationship trends that can be used to design and optimize future optical imaging probes. Furthermore, they confirm that a hydrophilic polymer is an ideal scaffold for use in optical imaging contrast probes, allowing a highly modular design that enables efficient optimization to maximize probe accumulation and overall biodistribution properties.
Collapse
Affiliation(s)
- Martin Hadzima
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo n. 2, Praha 6 16610, Czech Republic
- Department
of Organic Chemistry, Faculty of Science, Charles University, Albertov 6, Praha 2 12800, Czech Republic
| | - Franco F. Faucher
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Kristýna Blažková
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Joshua J. Yim
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Matteo Guerra
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Shiyu Chen
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Emily C. Woods
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Ki Wan Park
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| | - Pavel Šácha
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo n. 2, Praha 6 16610, Czech Republic
| | - Vladimír Šubr
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
n. 2, Praha 6 16206, Czech Republic
| | - Libor Kostka
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
n. 2, Praha 6 16206, Czech Republic
| | - Tomáš Etrych
- Institute
of Macromolecular Chemistry, Czech Academy
of Sciences, Heyrovského
n. 2, Praha 6 16206, Czech Republic
| | - Pavel Majer
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo n. 2, Praha 6 16610, Czech Republic
| | - Jan Konvalinka
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo n. 2, Praha 6 16610, Czech Republic
| | - Matthew Bogyo
- Department
of Pathology, School of Medicine, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
6
|
Roschelle M, Rabbani R, Gweon S, Kumar R, Vercruysse A, Cho NW, Spitzer MH, Niknejad AM, Stojanović VM, Anwar M. A Wireless, Multicolor Fluorescence Image Sensor Implant for Real-Time Monitoring in Cancer Therapy. ARXIV 2024:arXiv:2406.18881v1. [PMID: 38979489 PMCID: PMC11230517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Real-time monitoring of dynamic biological processes in the body is critical to understanding disease progression and treatment response. This data, for instance, can help address the lower than 50% response rates to cancer immunotherapy. However, current clinical imaging modalities lack the molecular contrast, resolution, and chronic usability for rapid and accurate response assessments. Here, we present a fully wireless image sensor featuring a 2.5×5 mm2 CMOS integrated circuit for multicolor fluorescence imaging deep in tissue. The sensor operates wirelessly via ultrasound (US) at 5 cm depth in oil, harvesting energy with 221 mW/cm2 incident US power density (31% of FDA limits) and backscattering data at 13 kbps with a bit error rate <10-6. In-situ fluorescence excitation is provided by micro-laser diodes controlled with a programmable on-chip driver. An optical frontend combining a multi-bandpass interference filter and a fiber optic plate provides >6 OD excitation blocking and enables three-color imaging for detecting multiple cell types. A 36×40-pixel array captures images with <125 μm resolution. We demonstrate wireless, dual-color fluorescence imaging of both effector and suppressor immune cells in ex vivo mouse tumor samples with and without immunotherapy. These results show promise for providing rapid insight into therapeutic response and resistance, guiding personalized medicine.
Collapse
Affiliation(s)
- Micah Roschelle
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley CA 94720 USA
| | - Rozhan Rabbani
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley CA 94720 USA
| | - Surin Gweon
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley CA 94720 USA
| | - Rohan Kumar
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley CA 94720 USA
| | - Alec Vercruysse
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley CA 94720 USA
| | - Nam Woo Cho
- Department of Radiation Oncology and the Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, CA 94158 USA
| | - Matthew H. Spitzer
- Department of Otolaryngology-Head and Neck Surgery and the Department of Microbiology and Immunology, University of California, San Francisco, CA 94158 USA
| | - Ali M. Niknejad
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley CA 94720 USA
| | - Vladimir M. Stojanović
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley CA 94720 USA
| | - Mekhail Anwar
- Department of Electrical Engineering and Computer Sciences, University of California at Berkeley, Berkeley, CA 94720 USA
- Department of Radiation Oncology, University of California, San Francisco, CA 94158 USA
| |
Collapse
|
7
|
Zhang Z, Du Y, Shi X, Wang K, Qu Q, Liang Q, Ma X, He K, Chi C, Tang J, Liu B, Ji J, Wang J, Dong J, Hu Z, Tian J. NIR-II light in clinical oncology: opportunities and challenges. Nat Rev Clin Oncol 2024; 21:449-467. [PMID: 38693335 DOI: 10.1038/s41571-024-00892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 05/03/2024]
Abstract
Novel strategies utilizing light in the second near-infrared region (NIR-II; 900-1,880 nm wavelengths) offer the potential to visualize and treat solid tumours with enhanced precision. Over the past few decades, numerous techniques leveraging NIR-II light have been developed with the aim of precisely eliminating tumours while maximally preserving organ function. During cancer surgery, NIR-II optical imaging enables the visualization of clinically occult lesions and surrounding vital structures with increased sensitivity and resolution, thereby enhancing surgical quality and improving patient prognosis. Furthermore, the use of NIR-II light promises to improve cancer phototherapy by enabling the selective delivery of increased therapeutic energy to tissues at greater depths. Initial clinical studies of NIR-II-based imaging and phototherapy have indicated impressive potential to decrease cancer recurrence, reduce complications and prolong survival. Despite the encouraging results achieved, clinical translation of innovative NIR-II techniques remains challenging and inefficient; multidisciplinary cooperation is necessary to bridge the gap between preclinical research and clinical practice, and thus accelerate the translation of technical advances into clinical benefits. In this Review, we summarize the available clinical data on NIR-II-based imaging and phototherapy, demonstrating the feasibility and utility of integrating these technologies into the treatment of cancer. We also introduce emerging NIR-II-based approaches with substantial potential to further enhance patient outcomes, while also highlighting the challenges associated with imminent clinical studies of these modalities.
Collapse
Affiliation(s)
- Zeyu Zhang
- Key Laboratory of Big Data-Based Precision Medicine of Ministry of Industry and Information Technology, School of Engineering Medicine, Beihang University, Beijing, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Xiaojing Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Qiaojun Qu
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Qian Liang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Xiaopeng Ma
- School of Control Science and Engineering, Shandong University, Jinan, China
| | - Kunshan He
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Chongwei Chi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China
| | - Jianqiang Tang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Liu
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiafu Ji
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, China.
| | - Jun Wang
- Thoracic Oncology Institute/Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China.
| | - Jiahong Dong
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China.
| | - Jie Tian
- Key Laboratory of Big Data-Based Precision Medicine of Ministry of Industry and Information Technology, School of Engineering Medicine, Beihang University, Beijing, China.
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, Beijing, China.
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China.
| |
Collapse
|
8
|
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
|
9
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.16.562247. [PMID: 37904924 PMCID: PMC10614810 DOI: 10.1101/2023.10.16.562247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
In curative-intent cancer surgery, intraoperative fluorescence imaging of both diseased and healthy tissue can help to ensure successful removal of all gross and microscopic disease 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 tumor and nerves in ex vivo prostate tissue.
Collapse
|
10
|
Kwon MJ, House BJ, Barth CW, Solanki A, Jones JA, Davis SC, Gibbs SL. Dual probe difference specimen imaging for prostate cancer margin assessment. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:082806. [PMID: 37082104 PMCID: PMC10111791 DOI: 10.1117/1.jbo.28.8.082806] [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: 07/15/2022] [Accepted: 03/31/2023] [Indexed: 05/03/2023]
Abstract
Significance Positive margin status due to incomplete removal of tumor tissue during radical prostatectomy for high-risk localized prostate cancer requires reoperation or adjuvant therapy, which increases morbidity and mortality. Adverse effects of prostate cancer treatments commonly include erectile dysfunction, urinary incontinence, and bowel dysfunction, making successful initial curative prostatectomy imperative. Aim Current intraoperative tumor margin assessment is largely limited to frozen section analysis, which is a lengthy, labor-intensive process that is obtrusive to the clinical workflow within the operating room (OR). Therefore, a rapid method for prostate cancer margin assessment in the OR could improve outcomes for patients. Approach Dual probe difference specimen imaging (DDSI), which uses paired antibody-based probes that are labeled with spectrally distinct fluorophores, was shown herein for prostate cancer margin assessment. The paired antibody-based probes consisted of a targeted probe to prostate-specific membrane antigen (PSMA) and an untargeted probe, which were used as a cocktail to stain resected murine tissue specimens including prostate tumor, adipose, muscle, and normal prostate. Ratiometric images (i.e., DDSI) of the difference between targeted and untargeted probe uptake were calculated and evaluated for accuracy using receiver operator characteristic curve analysis with area under the curve values used to evaluate the utility of the DDSI method to detect PSMA positive prostate cancer. Results Targeted and untargeted probe uptake was similar between the high and low PSMA expressing tumor due to nonspecific probe uptake after topical administration. The ratiometric DDSI approach showed substantial contrast difference between the PSMA positive tumors and their respective normal tissues (prostate, adipose, muscle). Furthermore, DDSI showed substantial contrast difference between the high PSMA expressing tumors and the minimally PSMA expressing tumors due to the ratiometric correction for the nonspecific uptake patterns in resected tissues. Conclusions Previous work has shown that ratiometic imaging has strong predictive value for breast cancer margin status using topical administration. Translation of the ratiometric DDSI methodology herein from breast to prostate cancers demonstrates it as a robust, ratiometric technique that provides a molecularly specific imaging modality for intraoperative margin detection. Using the validated DDSI protocol on resected prostate cancers permitted rapid and accurate assessment of PSMA status as a surrogate for prostate cancer margin status. Future studies will further evaluate the utility of this technology to quantitatively characterize prostate margin status using PSMA as a biomarker.
Collapse
Affiliation(s)
- Marcus J. Kwon
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Broderick J. House
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Connor W. Barth
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Allison Solanki
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Jocelyn A. Jones
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Scott C. Davis
- Thayer School of Engineering at Dartmouth College, Hanover, New Hampshire, United States
| | - Summer L. Gibbs
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States
- Address all correspondence to Summer L. Gibbs,
| |
Collapse
|
11
|
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: 5] [Impact Index Per Article: 5.0] [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
|
12
|
Chauhan N, Cabrera M, Chowdhury P, Nagesh PK, Dhasmana A, Pranav, Jaggi M, Chauhan SC, Yallapu MM. Indocyanine Green-based Glow Nanoparticles Probe for Cancer Imaging. Nanotheranostics 2023; 7:353-367. [PMID: 37151801 PMCID: PMC10161388 DOI: 10.7150/ntno.78405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 03/22/2023] [Indexed: 08/31/2023] Open
Abstract
Indocyanine green (ICG) is one of the FDA-approved near infra-red fluorescent (NIRF) probes for cancer imaging and image-guided surgery in the clinical setting. However, the limitations of ICG include poor photostability, high concentration toxicity, short circulation time, and poor cancer cell specificity. To overcome these hurdles, we engineered a nanoconstruct composed of poly (vinyl pyrrolidone) (PVP)-indocyanine green that is cloaked self-assembled with tannic acid (termed as indocyanine green-based glow nanoparticles probe, ICG-Glow NPs) for the cancer cell/tissue-specific targeting. The self-assembled ICG-Glow NPs were confirmed by spherical nanoparticles formation (DLS and TEM) and spectral analyses. The NIRF imaging characteristic of ICG-Glow NPs was established by superior fluorescence counts on filter paper and chicken tissue. The ICG-Glow NPs exhibited excellent hemo and cellular compatibility with human red blood cells, kidney normal, pancreatic normal, and other cancer cell lines. An enhanced cancer-specific NIRF binding and imaging capability of ICG-Glow NPs was confirmed using different human cancer cell lines and human tumor tissues. Additionally, tumor-specific binding/accumulation of ICG-Glow NPs was confirmed in MDA-MB-231 xenograft mouse model. Collectively, these findings suggest that ICG-Glow NPs have great potential as a novel and safe NIRF imaging probe for cancer cell/tumor imaging. This can lead to a quicker cancer diagnosis facilitating precise disease detection and management.
Collapse
Affiliation(s)
- Neeraj Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Marco Cabrera
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Prashanth K.B. Nagesh
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Pranav
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| | - Murali M. Yallapu
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, United States
| |
Collapse
|
13
|
Cheng Z, Jin Y, Li J, Shi G, Yu L, Shao B, Tian J, Du Y, Yuan Z. Fibronectin-targeting and metalloproteinase-activatable smart imaging probe for fluorescence imaging and image-guided surgery of breast cancer. J Nanobiotechnology 2023; 21:112. [PMID: 36978072 PMCID: PMC10053476 DOI: 10.1186/s12951-023-01868-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Residual lesions in the tumor bed have been a challenge for conventional white-light breast-conserving surgery. Meanwhile, lung micro-metastasis also requires improved detection methods. Intraoperative accurate identification and elimination of microscopic cancer can improve surgery prognosis. In this study, a smart fibronectin-targeting and metalloproteinase-activatable imaging probe CREKA-GK8-QC is developed. CREKA-GK8-QC possesses an average diameter of 21.7 ± 2.5 nm, excellent MMP-9 protein responsiveness and no obvious cytotoxicity. In vivo experiments demonstrate that NIR-I fluorescence imaging of CREKA-GK8-QC precisely detects orthotopic breast cancer and micro-metastatic lesions (nearly 1 mm) of lungs with excellent imaging contrast ratio and spatial resolution. More notably, fluorescence image-guided surgery facilitates complete resection and avoids residual lesions in the tumor bed, improving survival outcomes. We envision that our newly developed imaging probe shows superior capacity for specific and sensitive targeted imaging, as well as providing guidance for accurate surgical resection of breast cancer.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yushen Jin
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing, 100013, China
| | - Jiaqian Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Guangyuan Shi
- University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Leyi Yu
- Haidian Section of Peking University Third Hospital, Beijing, 100080, China
| | - Bing Shao
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing, 100013, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine Science and Engineering, Beihang University, Beijing, 100191, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100080, China.
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China.
| |
Collapse
|
14
|
Cheng Z, Ma J, Yin L, Yu L, Yuan Z, Zhang B, Tian J, Du Y. Non-invasive molecular imaging for precision diagnosis of metastatic lymph nodes: opportunities from preclinical to clinical applications. Eur J Nucl Med Mol Imaging 2023; 50:1111-1133. [PMID: 36443568 DOI: 10.1007/s00259-022-06056-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/18/2022] [Indexed: 11/30/2022]
Abstract
Lymph node metastasis is an indicator of the invasiveness and aggressiveness of cancer. It is a vital prognostic factor in clinical staging of the disease and therapeutic decision-making. Patients with positive metastatic lymph nodes are likely to develop recurrent disease, distant metastasis, and succumb to death in the coming few years. Lymph node dissection and histological analysis are needed to detect whether regional lymph nodes have been infiltrated by cancer cells and determine the likely outcome of treatment and the patient's chances of survival. However, these procedures are invasive, and tissue biopsies are prone to sampling error. In recent years, advanced molecular imaging with novel imaging probes has provided new technologies that are contributing to comprehensive management of cancer, including non-invasive investigation of lymphatic drainage from tumors, identifying metastatic lymph nodes, and guiding surgeons to operate efficiently in patients with complex lesions. In this review, first, we outline the current status of different molecular imaging modalities applied for lymph node metastasis management. Second, we summarize the multi-functional imaging probes applied with the different imaging modalities as well as applications of cancer lymph node metastasis from preclinical studies to clinical translations. Third, we describe the limitations that must be considered in the field of molecular imaging for improved detection of lymph node metastasis. Finally, we propose future directions for molecular imaging technology that will allow more personalized treatment plans for patients with lymph node metastasis.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China.,CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiaojiao Ma
- Department of Medical Ultrasonics, China-Japan Friendship Hospital, Yinghua East Road 2#, ChaoYang Dist., Beijing, 100029, China
| | - Lin Yin
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Leyi Yu
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China.
| | - Bo Zhang
- Department of Medical Ultrasonics, China-Japan Friendship Hospital, Yinghua East Road 2#, ChaoYang Dist., Beijing, 100029, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China. .,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine Science and Engineering, Beihang University, Beijing, 100191, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China. .,University of Chinese Academy of Sciences, Beijing, 100080, China.
| |
Collapse
|
15
|
Mordini D, Mavridi-Printezi A, Menichetti A, Cantelli A, Li X, Montalti M. Luminescent Gold Nanoclusters for Bioimaging: Increasing the Ligand Complexity. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13040648. [PMID: 36839016 PMCID: PMC9960743 DOI: 10.3390/nano13040648] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 05/31/2023]
Abstract
Fluorescence, and more in general, photoluminescence (PL), presents important advantages for imaging with respect to other diagnostic techniques. In particular, detection methodologies exploiting fluorescence imaging are fast and versatile; make use of low-cost and simple instrumentations; and are taking advantage of newly developed powerful, low-cost, light-based electronic devices, such as light sources and cameras, used in huge market applications, such as civil illumination, computers, and cellular phones. Besides the aforementioned simplicity, fluorescence imaging offers a spatial and temporal resolution that can hardly be achieved with alternative methods. However, the two main limitations of fluorescence imaging for bio-application are still (i) the biological tissue transparency and autofluorescence and (ii) the biocompatibility of the contrast agents. Luminescent gold nanoclusters (AuNCs), if properly designed, combine high biocompatibility with PL in the near-infrared region (NIR), where the biological tissues exhibit higher transparency and negligible autofluorescence. However, the stabilization of these AuNCs requires the use of specific ligands that also affect their PL properties. The nature of the ligand plays a fundamental role in the development and sequential application of PL AuNCs as probes for bioimaging. Considering the importance of this, in this review, the most relevant and recent papers on AuNCs-based bioimaging are presented and discussed highlighting the different functionalities achieved by increasing the complexity of the ligand structure.
Collapse
|
16
|
Fushiki H, Yoshikawa T, Matsuda T, Sato T, Suwa A. Preclinical Development and Validation of ASP5354: A Near-Infrared Fluorescent Agent for Intraoperative Ureter Visualization. Mol Imaging Biol 2023; 25:74-84. [PMID: 33977418 DOI: 10.1007/s11307-021-01613-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/31/2021] [Accepted: 04/28/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE Iatrogenic ureteral injury (IUI) can complicate minimally invasive and open abdominopelvic surgery. The incidence of IUI is low and dependent on the type of surgery (< 10 %), but it is associated with high morbidity. Therefore, intraoperative visualization of the ureter is critical to reduce the incidence of IUI, and some methodologies for ureter visualization have been developed. Amongst these, near-infrared fluorescence (NIRF) visualization is thought to bring an advantage with real-time retroperitoneal visualization through the retroperitoneum. We investigated an indocyanine green (ICG) derivative, ASP5354, which emits NIRF at 820 nm when exposed to near-infrared light at a wavelength of 780 nm, in a rodent and porcine model. PROCEDURES Wistar rats and Göttingen minipigs under anesthesia were laparotomized and then administered ASP5354 chloride intravenously at dose of 0.03 and 0.3 mg/kg for rats and 0.001 and 0.01 mg/kg for minipigs, respectively. Videos of the abdominal cavity in minipigs were taken using a near-infrared fluorescent camera (pde-neo) and assessed visually by three independent clinicians. Toxicological evaluation was demonstrated with cynomolgus monkeys. RESULTS The proportion of animals whose ureters were visible up to 3 h after administration of ASP5354 chloride were 33 % at 0.001 mg/kg and 100 % at 0.01 mg/kg, respectively. In a toxicological study in cynomolgus monkeys, ASP5354 chloride demonstrated no significant toxicity, suggesting that 0.01 mg/kg provides an optimal dose when used clinically and could allow for ureter visualization during routine surgical procedures. CONCLUSIONS The dose of 0.01 mg/kg provided an optimal dose for ureter visualization up to 3 h after administration. ASP5354 shows promise for ureter visualization during abdominopelvic surgery, which may potentially lower the risk of IUI.
Collapse
Affiliation(s)
- Hiroshi Fushiki
- Astellas Pharma, Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - Tomoaki Yoshikawa
- Astellas Pharma, Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - Toshihiro Matsuda
- Astellas Pharma, Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - Takeshi Sato
- Astellas Pharma, Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - Akira Suwa
- Astellas Pharma, Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan.
| |
Collapse
|
17
|
Li C, Torres VC, He Y, Xu X, Papavasiliou G, Samkoe KS, Brankov JG, Tichauer KM. Quantifying imaging agent binding and dissociation in 3D cancer spheroid tissue culture using paired-agent principles. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2023; 12360:123600L. [PMID: 37180093 PMCID: PMC10174639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Binding kinetics play an important role in cancer diagnosis and therapeutics. However, current methods of quantifying binding kinetics fail to consider the three-dimensional environment that drugs and imaging agents experience in biological tissue. In response, a methodology to assay agent binding and dissociation in 3D tissue culture was developed using paired-agent molecular imaging principles. To test the methodology, the uptakes of ABY-029 (an IRDye 800CW-labeled epidermal growth factor receptor (EGFR)-targeted antibody-mimetic) and IRDye 700DX-carboxylate in 3D spheroids were measured in four different human cancer cell lines throughout staining and rinsing. A compartment model (optimized for the application) was then fit to the kinetic curves of both imaging agents to estimate binding and dissociation rate constants of the EGFR targeted ABY-029 agent. A linear correlation was observed between apparent association rate constant (k 3 ) and the receptor concentration experimentally and in simulations (r = 0.99 , p < 0.05 ). Additionally, a similar binding affinity profile compared to a gold standard method was determined by this model. This low-cost methodology to quantify imaging agent or drug binding affinity in clinically relevant 3D tumor spheroid models, can be used to guide timing of imaging in molecular guided surgery and could have implications in drug development.
Collapse
Affiliation(s)
- Chengyue Li
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616
| | - Veronica C. Torres
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616
| | - Yusheng He
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616
| | - Xiaochun Xu
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756
| | | | - Kimberley S. Samkoe
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756
| | - Jovan G. Brankov
- Electrical and Computer Engineering, Illinois Institute of Technology, Chicago, IL, 60616
| | - Kenneth M. Tichauer
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, 60616
| |
Collapse
|
18
|
Quantum Dots Mediated Imaging and Phototherapy in Cancer Spheroid Models: State of the Art and Perspectives. Pharmaceutics 2022; 14:pharmaceutics14102136. [PMID: 36297571 PMCID: PMC9611360 DOI: 10.3390/pharmaceutics14102136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
Quantum Dots (QDs) are fluorescent nanoparticles known for their exceptional optical properties, i.e., high fluorescence emission, photostability, narrow emission spectrum, and broad excitation wavelength. These properties make QDs an exciting choice for bioimaging applications, notably in cancer imaging. Challenges lie in their ability to specifically label targeted cells. Numerous studies have been carried out with QDs coupled to various ligands like peptides, antibodies, aptamers, etc., to achieve efficient targeting. Most studies were conducted in vitro with two-dimensional cell monolayers (n = 8902) before evolving towards more sophisticated models. Three-dimensional multicellular tumor models better recapitulate in vivo conditions by mimicking cell-to-cell and cell-matrix interactions. To date, only few studies (n = 34) were conducted in 3D in vitro models such as spheroids, whereas these models could better represent QDs behavior in tumors compared to monolayers. Thus, the purpose of this review is to present a state of the art on the studies conducted with Quantum Dots on spheroid models for imaging and phototherapy purposes.
Collapse
|
19
|
Wang H, Kang H, Dinh J, Yokomizo S, Stiles WR, Tully M, Cardenas K, Srinivas S, Ingerick J, Ahn S, Bao K, Choi HS. P800SO3-PEG: a renal clearable bone-targeted fluorophore for theranostic imaging. Biomater Res 2022; 26:51. [PMID: 36183117 PMCID: PMC9526902 DOI: 10.1186/s40824-022-00294-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/31/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Due to the deep tissue penetration and reduced scattering, NIR-II fluorescence imaging is advantageous over conventional visible and NIR-I fluorescence imaging for the detection of bone growth, metabolism, metastasis, and other bone-related diseases. METHODS Bone-targeted heptamethine cyanine fluorophores were synthesized by substituting the meso-carbon with a sulfur atom, resulting in a bathochromic shift and increased fluorescence intensity. The physicochemical, optical, and thermal stability of newly synthesized bone-targeted NIR fluorophores was performed in aqueous solvents. Calcium binding, bone-specific targeting, biodistribution, pharmacokinetics, and 2D and 3D NIR imaging were performed in animal models. RESULTS The newly synthesized S-substituted heptamethine fluorophores demonstrated a high affinity for hydroxyapatite and calcium phosphate, which improved bone-specific targeting with signal-background ratios > 3.5. Particularly, P800SO3-PEG showed minimum nonspecific uptake, and most unbound molecules were excreted into the urinary bladder. Histological analyses demonstrated that P800SO3-PEG remained stable in the bone for over two weeks and was incorporated into bone matrices. Interestingly, the flexible thiol ethylene glycol linker on P800SO3-PEG induced a promising photothermal effect upon NIR laser irradiation, demonstrating potential theranostic imaging. CONCLUSIONS P800SO3-PEG shows a high affinity for bone tissues, deeper tissue imaging capabilities, minimum nonspecific uptake in the major organs, and photothermal effect upon laser irradiation, making it optimal for bone-targeted theranostic imaging.
Collapse
Affiliation(s)
- Haoran Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China.,Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jason Dinh
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shinya Yokomizo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wesley R Stiles
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Molly Tully
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kevin Cardenas
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Surbhi Srinivas
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jason Ingerick
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sung Ahn
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kai Bao
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Pace J, Ivich F, Marple E, Niedre M. Near-infrared diffuse in vivo flow cytometry. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-220101GR. [PMID: 36114606 PMCID: PMC9478904 DOI: 10.1117/1.jbo.27.9.097002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/07/2022] [Indexed: 06/15/2023]
Abstract
Significance Diffuse in vivo flow cytometry (DiFC) is an emerging technique for enumerating rare fluorescently labeled circulating cells noninvasively in the bloodstream. Thus far, we have reported red and blue-green versions of DiFC. Use of near-infrared (NIR) fluorescent light would in principle allow use of DiFC in deeper tissues and would be compatible with emerging NIR fluorescence molecular contrast agents. Aim We describe the design of a NIR-DiFC instrument and demonstrate its use in optical flow phantoms in vitro and in mice in vivo. Approach We developed an improved optical fiber probe design for efficient collection of fluorescence from individual circulating cells and efficient rejection of instrument autofluorescence. We built a NIR-DiFC instrument. We tested this with NIR fluorescent microspheres and cell lines labeled with OTL38 fluorescence contrast agent in a flow phantom model. We also tested NIR-DiFC in nude mice injected intravenously with OTL38-labeled L1210A cells. Results NIR-DiFC allowed detection of circulating tumor cells (CTCs) in flow phantoms with mean signal-to-noise ratios (SNRs) of 19 to 32 dB. In mice, fluorescently labeled CTCs were detectable with mean SNR of 26 dB. NIR-DiFC also exhibited orders significantly lower autofluorescence and false-alarm rates than blue-green DiFC. Conclusions NIR-DiFC allows use of emerging NIR contrast agents. Our work could pave the way for future use of NIR-DiFC in humans.
Collapse
Affiliation(s)
- Joshua Pace
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Fernando Ivich
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Eric Marple
- EmVision LLC, Loxahatchee, Florida, United States
| | - Mark Niedre
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| |
Collapse
|
21
|
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: 0.5] [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
|
22
|
Mander S, Naffouje SA, Gao J, Li W, Christov K, Green A, Bongarzone ER, Das Gupta TK, Yamada T. Tumor-targeting cell-penetrating peptide, p28, for glioblastoma imaging and therapy. Front Oncol 2022; 12:940001. [PMID: 35936749 PMCID: PMC9353713 DOI: 10.3389/fonc.2022.940001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Despite recent advances in cancer research, glioblastoma multiforme (GBM) remains a highly aggressive brain tumor as its treatment options are limited. The current standard treatment includes surgery followed by radiotherapy and adjuvant chemotherapy. However, surgery without image guidance is often challenging to achieve maximal safe resection as it is difficult to precisely discern the lesion to be removed from surrounding brain tissue. In addition, the efficacy of adjuvant chemotherapy is limited by poor penetration of therapeutics through the blood-brain barrier (BBB) into brain tissues, and the lack of tumor targeting. In this regard, we utilized a tumor-targeting cell-penetration peptide, p28, as a therapeutic agent to improve the efficacy of a current chemotherapeutic agent for GBM, and as a carrier for a fluorescence imaging agent for a clear identification of GBM. Here, we show that a near-infrared (NIR) imaging agent, ICG-p28 (a chemical conjugate of an FDA-approved NIR dye, indocyanine green ICG, and tumor-targeting p28 peptide) can preferentially localize tumors in multiple GBM animal models. Moreover, xenograft studies show that p28, as a therapeutic agent, can enhance the cytotoxic activity of temozolomide (TMZ), one of the few effective drugs for brain tumors. Collectively, our findings highlight the important role of the tumor-targeting peptide, which has great potential for intraoperative image-guided surgery and the development of new therapeutic strategies for GBM.
Collapse
Affiliation(s)
- Sunam Mander
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL, United States
| | - Samer A. Naffouje
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL, United States
| | - Jin Gao
- Department of Electrical and Computer Engineering, University of Illinois College of Engineering, Chicago, IL, United States
| | - Weiguo Li
- Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Engineering, Chicago, IL, United States
| | - Konstantin Christov
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL, United States
| | - Albert Green
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL, United States
| | - Ernesto R. Bongarzone
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, Chicago, IL, United States
| | - Tapas K. Das Gupta
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL, United States
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL, United States
- Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Engineering, Chicago, IL, United States
- *Correspondence: Tohru Yamada,
| |
Collapse
|
23
|
Yokomizo S, Henary M, Buabeng ER, Fukuda T, Monaco H, Baek Y, Manganiello S, Wang H, Kubota J, Ulumben AD, Lv X, Wang C, Inoue K, Fukushi M, Kang H, Bao K, Kashiwagi S, Choi HS. Topical pH Sensing NIR Fluorophores for Intraoperative Imaging and Surgery of Disseminated Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201416. [PMID: 35567348 PMCID: PMC9286000 DOI: 10.1002/advs.202201416] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Indexed: 05/05/2023]
Abstract
Fluorescence-guided surgery (FGS) aids surgeons with real-time visualization of small cancer foci and borders, which improves surgical and prognostic efficacy of cancer. Despite the steady advances in imaging devices, there is a scarcity of fluorophores available to achieve optimal FGS. Here, 1) a pH-sensitive near-infrared fluorophore that exhibits rapid signal changes in acidic tumor microenvironments (TME) caused by the attenuation of intramolecular quenching, 2) the inherent targeting for cancer based on chemical structure (structure inherent targeting, SIT), and 3) mitochondrial and lysosomal retention are reported. After topical application of PH08 on peritoneal tumor regions in ovarian cancer-bearing mice, a rapid fluorescence increase (< 10 min), and extended preservation of signals (> 4 h post-topical application) are observed, which together allow for the visualization of submillimeter tumors with a high tumor-to-background ratio (TBR > 5.0). In addition, PH08 is preferentially transported to cancer cells via organic anion transporter peptides (OATPs) and colocalizes in the mitochondria and lysosomes due to the positive charges, enabling a long retention time during FGS. PH08 not only has a significant impact on surgical and diagnostic applications but also provides an effective and scalable strategy to design therapeutic agents for a wide array of cancers.
Collapse
Affiliation(s)
- Shinya Yokomizo
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
- Department of Radiological SciencesTokyo Metropolitan University7‐2‐10 Higashi‐OguArakawaTokyo116–8551Japan
| | - Maged Henary
- Department of Chemistry and Center for Diagnostics and TherapeuticsGeorgia State University100 Piedmont Avenue SEAtlantaGA30303USA
| | - Emmanuel R. Buabeng
- Department of Chemistry and Center for Diagnostics and TherapeuticsGeorgia State University100 Piedmont Avenue SEAtlantaGA30303USA
| | - Takeshi Fukuda
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
- Department of Obstetrics and GynecologyOsaka City University Graduate School of Medicine1‐4‐3, AsahimachiAbeno‐kuOsaka545–8585Japan
| | - Hailey Monaco
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Yoonji Baek
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Sophia Manganiello
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Haoran Wang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Jo Kubota
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Amy Daniel Ulumben
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Xiangmin Lv
- Vincent Center for Reproductive BiologyVincent Department of Obstetrics and GynecologyMassachusetts General HospitalBostonMA02114USA
| | - Cheng Wang
- Vincent Center for Reproductive BiologyVincent Department of Obstetrics and GynecologyMassachusetts General HospitalBostonMA02114USA
| | - Kazumasa Inoue
- Department of Radiological SciencesTokyo Metropolitan University7‐2‐10 Higashi‐OguArakawaTokyo116–8551Japan
| | - Masahiro Fukushi
- Department of Radiological SciencesTokyo Metropolitan University7‐2‐10 Higashi‐OguArakawaTokyo116–8551Japan
| | - Homan Kang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Kai Bao
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Hak Soo Choi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| |
Collapse
|
24
|
Leiloglou M, Kedrzycki MS, Chalau V, Chiarini N, Thiruchelvam PTR, Hadjiminas DJ, Hogben KR, Rashid F, Ramakrishnan R, Darzi AW, Leff DR, Elson DS. Indocyanine green fluorescence image processing techniques for breast cancer macroscopic demarcation. Sci Rep 2022; 12:8607. [PMID: 35597783 PMCID: PMC9124184 DOI: 10.1038/s41598-022-12504-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/05/2022] [Indexed: 12/13/2022] Open
Abstract
Re-operation due to disease being inadvertently close to the resection margin is a major challenge in breast conserving surgery (BCS). Indocyanine green (ICG) fluorescence imaging could be used to visualize the tumor boundaries and help surgeons resect disease more efficiently. In this work, ICG fluorescence and color images were acquired with a custom-built camera system from 40 patients treated with BCS. Images were acquired from the tumor in-situ, surgical cavity post-excision, freshly excised tumor and histopathology tumour grossing. Fluorescence image intensity and texture were used as individual or combined predictors in both logistic regression (LR) and support vector machine models to predict the tumor extent. ICG fluorescence spectra in formalin-fixed histopathology grossing tumor were acquired and analyzed. Our results showed that ICG remains in the tissue after formalin fixation. Therefore, tissue imaging could be validated in freshly excised and in formalin-fixed grossing tumor. The trained LR model with combined fluorescence intensity (pixel values) and texture (slope of power spectral density curve) identified the tumor's extent in the grossing images with pixel-level resolution and sensitivity, specificity of 0.75 ± 0.3, 0.89 ± 0.2.This model was applied on tumor in-situ and surgical cavity (post-excision) images to predict tumor presence.
Collapse
Affiliation(s)
- Maria Leiloglou
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK. .,Department of Surgery and Cancer, Imperial College London, London, UK.
| | - Martha S Kedrzycki
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK.,Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Vadzim Chalau
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| | - Nicolas Chiarini
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Paul T R Thiruchelvam
- Department of Surgery and Cancer, Imperial College London, London, UK.,Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Dimitri J Hadjiminas
- Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Katy R Hogben
- Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Faiza Rashid
- Department of Histopathology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Rathi Ramakrishnan
- Department of Histopathology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Ara W Darzi
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| | - Daniel R Leff
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK.,Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Daniel S Elson
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
25
|
Niedre M. Prospects for Fluorescence Molecular In Vivo Liquid Biopsy of Circulating Tumor Cells in Humans. FRONTIERS IN PHOTONICS 2022; 3:910035. [PMID: 39508030 PMCID: PMC11540420 DOI: 10.3389/fphot.2022.910035] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Our team recently developed "Diffuse in vivo Flow Cytometry" (DiFC) for detection and enumeration rare circulating tumor cells (CTCs) in mice with highly-scattered fluorescent light. We have used DiFC to study dissemination of CTCs in a number of mouse models of metastasis with fluorescent protein expressing cells. Because DiFC uses diffuse light and interrogates large blood vessels in relatively deep tissue, in principle it could be translated to larger limbs, species, and even humans clinically. In this perspective, we discuss the technical challenges of human translation of DiFC in the context of the current state of the technology, as well as potential strategies for labeling of CTCs with targeted fluorescent molecular probes. We also discuss potential advantages and disadvantages of DiFC as a clinical tool. In principle, DiFC could represent a powerful complementary technique (to liquid biopsy blood draws) for accurate and sensitive measurement of changes in CTC numbers over time.
Collapse
Affiliation(s)
- Mark Niedre
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| |
Collapse
|
26
|
Litorja M. Conversion of imager-specific response to tissue phantom fluorescence into system of units-traceable units. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-210303SSRR. [PMID: 35552461 PMCID: PMC9098210 DOI: 10.1117/1.jbo.27.7.074715] [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: 10/12/2021] [Accepted: 04/14/2022] [Indexed: 06/15/2023]
Abstract
SIGNIFICANCE The fluorescence-guided imaging for surgical intervention community recognizes the need for performance standards for these imaging devices. Tissue phantoms are used to track an imager's performance as a fluorescence detector, but imager-specific units are of limited utility. AIM Tissue phantoms can be calibrated to be traceable to the international system of units (SI) and in turn be used to calibrate imagers such that fluorescence measurements can be reported in universally accepted units. APPROACH The radiometry to convert imager-specific arbitrary digital counts to SI-traceable unit of watts is described in this paper. RESULTS An example of an imager calibration is included. CONCLUSIONS Calibrated tissue phantoms become a tool for metrological traceability.
Collapse
Affiliation(s)
- Maritoni Litorja
- National Institute of Standards and Technology, Sensor Science Division, Gaithersburg, Maryland, United States
| |
Collapse
|
27
|
Targeted multicolor in vivo imaging over 1,000 nm enabled by nonamethine cyanines. Nat Methods 2022; 19:353-358. [PMID: 35228725 DOI: 10.1038/s41592-022-01394-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/05/2022] [Indexed: 02/07/2023]
Abstract
Recent progress has shown that using wavelengths between 1,000 and 2,000 nm, referred to as the shortwave-infrared or near-infrared (NIR)-II range, can enable high-resolution in vivo imaging at depths not possible with conventional optical wavelengths. However, few bioconjugatable probes of the type that have proven invaluable for multiplexed imaging in the visible and NIR range are available for imaging these wavelengths. Using rational design, we have generated persulfonated indocyanine dyes with absorbance maxima at 872 and 1,072 nm through catechol-ring and aryl-ring fusion, respectively, onto the nonamethine scaffold. Multiplexed two-color and three-color in vivo imaging using monoclonal antibody and dextran conjugates in several tumor models illustrate the benefits of concurrent labeling of the tumor and healthy surrounding tissue and lymphatics. These efforts are enabled by complementary advances in a custom-built NIR/shortwave-infrared imaging setup and software package for multicolor real-time imaging.
Collapse
|
28
|
Zhang Z, He K, Chi C, Hu Z, Tian J. Intraoperative fluorescence molecular imaging accelerates the coming of precision surgery in China. Eur J Nucl Med Mol Imaging 2022; 49:2531-2543. [PMID: 35230491 PMCID: PMC9206608 DOI: 10.1007/s00259-022-05730-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/13/2022] [Indexed: 02/06/2023]
Abstract
Purpose China has the largest cancer population globally. Surgery is the main choice for most solid cancer patients. Intraoperative fluorescence molecular imaging (FMI) has shown its great potential in assisting surgeons in achieving precise resection. We summarized the typical applications of intraoperative FMI and several new trends to promote the development of precision surgery. Methods The academic database and NIH clinical trial platform were systematically evaluated. We focused on the clinical application of intraoperative FMI in China. Special emphasis was placed on a series of typical studies with new technologies or high-level evidence. The emerging strategy of combining FMI with other modalities was also discussed. Results The clinical applications of clinically approved indocyanine green (ICG), methylene blue (MB), or fluorescein are on the rise in different surgical departments. Intraoperative FMI has achieved precise lesion detection, sentinel lymph node mapping, and lymphangiography for many cancers. Nerve imaging is also exploring to reduce iatrogenic injuries. Through different administration routes, these fluorescent imaging agents provided encouraging results in surgical navigation. Meanwhile, designing new cancer-specific fluorescent tracers is expected to be a promising trend to further improve the surgical outcome. Conclusions Intraoperative FMI is in a rapid development in China. In-depth understanding of cancer-related molecular mechanisms is necessary to achieve precision surgery. Molecular-targeted fluorescent agents and multi-modal imaging techniques might play crucial roles in the era of precision surgery.
Collapse
Affiliation(s)
- Zeyu Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China.,CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Kunshan He
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,State Key Laboratory of Computer Science and Beijing Key Lab of Human-Computer Interaction, Institute of Software, Chinese Academy of Sciences, Beijing, China
| | - Chongwei Chi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China.
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China.
| | - Jie Tian
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China. .,CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
29
|
Michie MS, Xu B, Sudlow G, Springer LE, Pham CT, Achilefu S. Side-chain modification of collagen-targeting peptide prevents dye aggregation for improved molecular imaging of arthritic joints. J Photochem Photobiol A Chem 2022; 424:113624. [PMID: 36406204 PMCID: PMC9673490 DOI: 10.1016/j.jphotochem.2021.113624] [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] [Indexed: 02/03/2023]
Abstract
Near-infrared (NIR) dye-peptide conjugates are widely used for tissue-targeted molecular fluorescence imaging of pathophysiologic conditions. However, the significant contribution of both dye and peptide to the net mass of these bioconjugates implies that small changes in either component could alter their photophysical and biological properties. Here, we synthesized and conjugated a type I collagen targeted peptide, RRANAALKAGELYKCILY, to either a hydrophobic (LS1000) or hydrophilic (LS1006) NIR fluorescent dye. Spectroscopic analysis revealed rapid self-assembly of both LS1000 and LS1006 in aqueous media to form stable dimeric/H aggregates, regardless of the free dye's solubility in water. We discovered that replacing the cysteine residue in LS1000 and LS1006 with acetamidomethyl cysteine to afford LS1001 and LS1107, respectively, disrupted the peptide's self-assembly and activated the previously quenched dye's fluorescence in aqueous conditions. These results highlight the dominant role of the octadecapeptide, but not the dye molecules, in controlling the photophysical properties of these conjugates by likely sequestering or extruding the hydrophobic or hydrophilic dyes, respectively. Application of the compounds for imaging collagen-rich tissue in an animal model of inflammatory arthritis showed enhanced uptake of all four conjugates, which retained high collagen-binding affinity, in inflamed joints. Moreover, LS1001 and LS1107 improved the arthritic joint-to-background contrast, suggesting that reduced aggregation enhanced the clearance of these compounds from non-target tissues. Our results highlight a peptide-driven strategy to alter the aggregation states of molecular probes in aqueous solutions, irrespective of the water-solubilizing properties of the dye molecules. The interplay between the monomeric and aggregated forms of the conjugates using simple thiol-modifiers lends the peptide-driven approach to diverse applications, including the effective imaging of inflammatory arthritis joints.
Collapse
Affiliation(s)
- Megan S. Michie
- Optical Radiology Laboratory, Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Baogang Xu
- Optical Radiology Laboratory, Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Gail Sudlow
- Optical Radiology Laboratory, Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Luke E. Springer
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Christine T.N. Pham
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Samuel Achilefu
- Optical Radiology Laboratory, Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
30
|
Saad MA, Xavierselvan M, Sharif HA, Selfridge S, Pawle R, Varvares M, Mallidi S, Hasan T. Dual Function Antibody Conjugates for Multimodal Imaging and Photoimmunotherapy of Cancer Cells. Photochem Photobiol 2022; 98:220-231. [PMID: 34379796 PMCID: PMC10038131 DOI: 10.1111/php.13501] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/08/2021] [Indexed: 11/29/2022]
Abstract
Precision imaging, utilizing molecular targeted agents, is an important tool in cancer diagnostics and guiding therapies. While there are limitations associated with single mode imaging probes, multimodal molecular imaging probes enabling target visualization through complementary imaging technologies provides an attractive alternative. However, there are several challenges associated with designing molecular probes carrying contrast agents for complementary multimodal imaging. Here, we propose a dual function antibody conjugate (DFAC) comprising an FDA approved photosensitizer Benzoporphyrin derivative (BPD) and a naphthalocyanine-based photoacoustic dye (SiNc(OH)) for multimodal infrared (IR) imaging. While fluorescence imaging, through BPD, provides sensitivity, complementing it with photoacoustic imaging, through SiNc(OH), provides a depth-resolved spatial resolution much beyond the optical diffusion limits of fluorescence measurements. Through a series of in vitro experiments, we demonstrate the development and utilization of DFACs for multimodal imaging and photodynamic treatment of squamous cell carcinoma (A431) cell line. The proposed DFACs have potential use in precision imaging applications such as guiding tumor resection surgeries and photodynamic treatment of residual microscopic disease thereby minimizing local recurrence. The data demonstrated in this study merits further investigation for its preclinical and clinical translation.
Collapse
Affiliation(s)
- Mohammad A. Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Marvin Xavierselvan
- Department of Biomedical Engineering, Science and Technology Center, Tufts University, Medford, MA
| | | | | | | | - Mark Varvares
- Department of Otolaryngology Head and Neck Surgery, Harvard Medical School, The Massachusetts Eye and Ear, Boston, MA
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Department of Biomedical Engineering, Science and Technology Center, Tufts University, Medford, MA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
31
|
Rijs Z, Jeremiasse B, Shifai N, Gelderblom H, Sier CFM, Vahrmeijer AL, van Leeuwen FWB, van der Steeg AFW, van de Sande MAJ. Introducing Fluorescence-Guided Surgery for Pediatric Ewing, Osteo-, and Rhabdomyosarcomas: A Literature Review. Biomedicines 2021; 9:biomedicines9101388. [PMID: 34680505 PMCID: PMC8533294 DOI: 10.3390/biomedicines9101388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023] Open
Abstract
Sarcomas are a rare heterogeneous group of malignant neoplasms of mesenchymal origin which represent approximately 13% of all cancers in pediatric patients. The most prevalent pediatric bone sarcomas are osteosarcoma (OS) and Ewing sarcoma (ES). Rhabdomyosarcoma (RMS) is the most frequently occurring pediatric soft tissue sarcoma. The median age of OS and ES is approximately 17 years, so this disease is also commonly seen in adults while non-pleiomorphic RMS is rare in the adult population. The mainstay of all treatment regimens is multimodal treatment containing chemotherapy, surgical resection, and sometimes (neo)adjuvant radiotherapy. A clear resection margin improves both local control and overall survival and should be the goal during surgery with a curative intent. Real-time intraoperative fluorescence-guided imaging could facilitate complete resections by visualizing tumor tissue during surgery. This review evaluates whether non-targeted and targeted fluorescence-guided surgery (FGS) could be beneficial for pediatric OS, ES, and RMS patients. Necessities for clinical implementation, current literature, and the positive as well as negative aspects of non-targeted FGS using the NIR dye Indocyanine Green (ICG) were evaluated. In addition, we provide an overview of targets that could potentially be used for FGS in OS, ES, and RMS. Then, due to the time- and cost-efficient translational perspective, we elaborate on the use of antibody-based tracers as well as their disadvantages and alternatives. Finally, we conclude with recommendations for the experiments needed before FGS can be implemented for pediatric OS, ES, and RMS patients.
Collapse
Affiliation(s)
- Zeger Rijs
- Department of Orthopedic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.S.); (M.A.J.v.d.S.)
- Correspondence: ; Tel.: +31-641-637-074
| | - Bernadette Jeremiasse
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (B.J.); (A.F.W.v.d.S.)
| | - Naweed Shifai
- Department of Orthopedic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.S.); (M.A.J.v.d.S.)
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands;
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (C.F.M.S.); (A.L.V.)
- Percuros BV, 2333 CL Leiden, The Netherlands
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (C.F.M.S.); (A.L.V.)
| | - Fijs W. B. van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands;
| | - Alida F. W. van der Steeg
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (B.J.); (A.F.W.v.d.S.)
| | - Michiel A. J. van de Sande
- Department of Orthopedic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.S.); (M.A.J.v.d.S.)
| |
Collapse
|
32
|
Kedrzycki MS, Leiloglou M, Chalau V, Chiarini N, Thiruchelvam PTR, Hadjiminas DJ, Hogben KR, Rashid F, Ramakrishnan R, Darzi AW, Elson DS, Leff DR. The Impact of Temporal Variation in Indocyanine Green Administration on Tumor Identification During Fluorescence Guided Breast Surgery. Ann Surg Oncol 2021; 28:5617-5625. [PMID: 34347221 PMCID: PMC8418597 DOI: 10.1245/s10434-021-10503-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/07/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND On average, 21% of women in the USA treated with Breast Conserving Surgery (BCS) undergo a second operation because of close positive margins. Tumor identification with fluorescence imaging could improve positive margin rates through demarcating location, size, and invasiveness of tumors. We investigated the technique's diagnostic accuracy in detecting tumors during BCS using intravenous indocyanine green (ICG) and a custom-built fluorescence camera system. METHODS In this single-center prospective clinical study, 40 recruited BCS patients were sub-categorized into two cohorts. In the first 'enhanced permeability and retention' (EPR) cohort, 0.25 mg/kg ICG was injected ~ 25 min prior to tumor excision, and in the second 'angiography' cohort, ~ 5 min prior to tumor excision. Subsequently, an in-house imaging system was used to image the tumor in situ prior to resection, ex vivo following resection, the resection bed, and during grossing in the histopathology laboratory to compare the technique's diagnostic accuracy between the cohorts. RESULTS The two cohorts were matched in patient and tumor characteristics. The majority of patients had invasive ductal carcinoma with concomitant ductal carcinoma in situ. Tumor-to-background ratio (TBR) in the angiography cohort was superior to the EPR cohort (TBR = 3.18 ± 1.74 vs 2.10 ± 0.92 respectively, p = 0.023). Tumor detection reached sensitivity and specificity scores of 0.82 and 0.93 for the angiography cohort and 0.66 and 0.90 for the EPR cohort, respectively (p = 0.1051 and p = 0.9099). DISCUSSION ICG administration timing during the angiography phase compared with the EPR phase improved TBR and diagnostic accuracy. Future work will focus on image pattern analysis and adaptation of the camera system to targeting fluorophores specific to breast cancer.
Collapse
Affiliation(s)
- Martha S Kedrzycki
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK.,Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Maria Leiloglou
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK. .,Department of Surgery and Cancer, Imperial College London, London, UK.
| | - Vadzim Chalau
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| | - Nicolas Chiarini
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Paul T R Thiruchelvam
- Department of Surgery and Cancer, Imperial College London, London, UK.,Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Dimitri J Hadjiminas
- Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Katy R Hogben
- Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Faiza Rashid
- Department of Histopathology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Rathi Ramakrishnan
- Department of Histopathology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Ara W Darzi
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| | - Daniel S Elson
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK
| | - Daniel R Leff
- Hamlyn Centre, Institute of Global Health Innovation, Imperial College London, London, UK.,Department of Surgery and Cancer, Imperial College London, London, UK.,Department of Breast Surgery, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
33
|
Lee YJ, van den Berg NS, Orosco RK, Rosenthal EL, Sorger JM. A narrative review of fluorescence imaging in robotic-assisted surgery. LAPAROSCOPIC SURGERY 2021; 5. [PMID: 34549180 PMCID: PMC8452263 DOI: 10.21037/ls-20-98] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Objective In this review, we provide examples of applications of fluorescence imaging in urologic, gynecologic, general, and endocrine surgeries. Background While robotic-assisted surgery has helped increase the availability of minimally invasive procedures across surgical specialties, there remains an opportunity to reduce adverse events associated with open, laparoscopic, and robotic-assisted methods. In 2011, fluorescence imaging was introduced as an option to the da Vinci Surgical System, and has been standard equipment since 2014. Without interfering with surgical workflow, this fluorescence technology named Firefly® allows for acquisition and display of near-infrared fluorescent signals that are co-registered with white light endoscopic images. As a result, robotic surgeons of all specialties have been able to explore the clinical utility of fluorescence guided surgery. Methods Literature searches were performed using the PubMed and MEDLINE databases using the keywords "robotic-assisted fluorescence surgery", "ICG robotic surgery", and "fluorescence guided surgery" covering the years 2011-2020. Conclusions Real-time intraoperative fluorescence guidance has shown great potential in helping guide surgeons in both simple and complex surgical interventions. Indocyanine green is one of the most widely-used imaging agents in fluorescence guided surgery, and other targeted, near-infrared imaging agents are in various stages of development. Fluorescence is becoming a reliable tool that can help surgeons in their decision-making process in some specialties, while explorations continue in others.
Collapse
Affiliation(s)
- Yu-Jin Lee
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Palo Alto, CA, USA
| | | | - Ryan K Orosco
- Moores Cancer Center, La Jolla, CA, USA.,Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of California, San Diego, San Diego, CA, USA
| | - Eben L Rosenthal
- Department of Otolaryngology-Head and Neck Surgery, Stanford University, Palo Alto, CA, USA
| | - Jonathan M Sorger
- Department of Research, Intuitive Surgical, Inc., Sunnyvale, CA, USA
| |
Collapse
|
34
|
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: 10.0] [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
|
35
|
Privat M, Bellaye PS, Lescure R, Massot A, Baffroy O, Moreau M, Racoeur C, Marcion G, Denat F, Bettaieb A, Collin B, Bodio E, Paul C, Goze C. Development of an Easily Bioconjugatable Water-Soluble Single-Photon Emission-Computed Tomography/Optical Imaging Bimodal Imaging Probe Based on the aza-BODIPY Fluorophore. J Med Chem 2021; 64:11063-11073. [PMID: 34338511 DOI: 10.1021/acs.jmedchem.1c00450] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A water-soluble fluorescent aza-BODIPY platform (Wazaby) was prepared and functionalized by a polyazamacrocycle agent and a bioconjugable arm. The resulting fluorescent derivative was characterized and bioconjugated onto a trastuzumab monoclonal antibody as a vector. After bioconjugation, the imaging agent appeared to be stable in serum (>72 h at 37 °C) and specifically labeled HER-2-positive breast tumors slices. The bioconjugate was radiolabeled with [111In] indium and studied in vivo. The developed monomolecular multimodal imaging probe (MOMIP) is water-soluble and chemically and photochemically stable, emits in the near infrared (NIR) region (734 nm in aqueous media), and displays a good quantum yield of fluorescence (around 15%). Single-photon emission-computed tomography and fluorescence imaging have been performed in nude mice bearing HER2-overexpressing HCC1954 human breast cancer xenografts and have evidenced the good tumor targeting of the [111In] In bimodal agent. Finally, the proof of concept of using it as a new tool for fluorescence-guided surgery has been shown.
Collapse
Affiliation(s)
- Malorie Privat
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France.,Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris; LIIC, EA7269, Université de Bourgogne, Franche Comté, Dijon 21000, France
| | - Pierre-Simon Bellaye
- Service de médecine nucléaire, Centre Georges François Leclerc, 1 rue Professeur Marion, BP77980, Dijon Cedex 21079, France
| | - Robin Lescure
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| | - Aurélie Massot
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris; LIIC, EA7269, Université de Bourgogne, Franche Comté, Dijon 21000, France
| | - Océane Baffroy
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| | - Mathieu Moreau
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France.,Service de médecine nucléaire, Centre Georges François Leclerc, 1 rue Professeur Marion, BP77980, Dijon Cedex 21079, France
| | - Cindy Racoeur
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris; LIIC, EA7269, Université de Bourgogne, Franche Comté, Dijon 21000, France
| | - Guillaume Marcion
- UMR INSERM/uB/AGROSUP 1231, Team 3 HSP-Pathies, labellisée Ligue Nationale contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France UFR des Sciences de Santé, Université de Bourgogne, Dijon 21000, France
| | - Franck Denat
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris; LIIC, EA7269, Université de Bourgogne, Franche Comté, Dijon 21000, France
| | - Bertrand Collin
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France.,Service de médecine nucléaire, Centre Georges François Leclerc, 1 rue Professeur Marion, BP77980, Dijon Cedex 21079, France
| | - Ewen Bodio
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris; LIIC, EA7269, Université de Bourgogne, Franche Comté, Dijon 21000, France
| | - Christine Goze
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| |
Collapse
|
36
|
Cantelli A, Malferrari M, Soldà A, Simonetti G, Forni S, Toscanella E, Mattioli EJ, Zerbetto F, Zanelli A, Di Giosia M, Zangoli M, Barbarella G, Rapino S, Di Maria F, Calvaresi M. Human Serum Albumin-Oligothiophene Bioconjugate: A Phototheranostic Platform for Localized Killing of Cancer Cells by Precise Light Activation. JACS AU 2021; 1:925-935. [PMID: 34467339 PMCID: PMC8395684 DOI: 10.1021/jacsau.1c00061] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 05/05/2023]
Abstract
The electronic, optical, and redox properties of thiophene-based materials have made them pivotal in nanoscience and nanotechnology. However, the exploitation of oligothiophenes in photodynamic therapy is hindered by their intrinsic hydrophobicity that lowers their biocompatibility and availability in water environments. Here, we developed human serum albumin (HSA)-oligothiophene bioconjugates that afford the use of insoluble oligothiophenes in physiological environments. UV-vis and electrophoresis proved the conjugation of the oligothiophene sensitizers to the protein. The bioconjugate is water-soluble and biocompatible, does not have any "dark toxicity", and preserves HSA in the physiological monomeric form, as confirmed by dynamic light scattering and circular dichroism measurements. In contrast, upon irradiation with ultralow light doses, the bioconjugate efficiently produces reactive oxygen species (ROS) and leads to the complete eradication of cancer cells. Real-time monitoring of the photokilling activity of the HSA-oligothiophene bioconjugate shows that living cells "explode" upon irradiation. Photodependent and dose-dependent apoptosis is one of the primary mechanisms of cell death activated by bioconjugate irradiation. The bioconjugate is a novel theranostic platform able to generate ROS intracellularly and provide imaging through the fluorescence of the oligothiophene. It is also a real-time self-reporting system able to monitor the apoptotic process. The induced phototoxicity is strongly confined to the irradiated region, showing localized killing of cancer cells by precise light activation of the bioconjugate.
Collapse
Affiliation(s)
- Andrea Cantelli
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Marco Malferrari
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Alice Soldà
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Giorgia Simonetti
- IRCCS
Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Via Piero Maroncelli, 40, 47014 Meldola, FC, Italy
| | - Sonny Forni
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Edoardo Toscanella
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Edoardo J. Mattioli
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Francesco Zerbetto
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Alberto Zanelli
- Istituto
per la Sintesi Organica e la Fotoreattività (ISOF), Consiglio Nazionale delle Ricerche, Via Piero Gobetti, 101, 40129 Bologna, Italy
| | - Matteo Di Giosia
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Mattia Zangoli
- Istituto
per la Sintesi Organica e la Fotoreattività (ISOF), Consiglio Nazionale delle Ricerche, Via Piero Gobetti, 101, 40129 Bologna, Italy
- Mediteknology
srl, Via Piero Gobetti,
101, 40129 Bologna, Italy
| | - Giovanna Barbarella
- Istituto
per la Sintesi Organica e la Fotoreattività (ISOF), Consiglio Nazionale delle Ricerche, Via Piero Gobetti, 101, 40129 Bologna, Italy
- Mediteknology
srl, Via Piero Gobetti,
101, 40129 Bologna, Italy
| | - Stefania Rapino
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| | - Francesca Di Maria
- Istituto
per la Sintesi Organica e la Fotoreattività (ISOF), Consiglio Nazionale delle Ricerche, Via Piero Gobetti, 101, 40129 Bologna, Italy
- Mediteknology
srl, Via Piero Gobetti,
101, 40129 Bologna, Italy
| | - Matteo Calvaresi
- Dipartimento
di Chimica “Giacomo Ciamician, Alma Mater Studiorum, Università di Bologna, Via Francesco Selmi, 2, 40126 Bologna, Italy
| |
Collapse
|
37
|
Abstract
Cysteine cathepsins are proteases critical in physiopathological processes and show potential as targets or biomarkers for diseases and medical conditions. The 11 members of the cathepsin family are redundant in some cases but remarkably independent of others, demanding the development of both pan-cathepsin targeting tools as well as probes that are selective for specific cathepsins with little off-target activity. This review addresses the diverse design strategies that have been employed to accomplish this tailored selectivity among cysteine cathepsin targets and the imaging modalities incorporated. The power of these diverse tools is contextualized by briefly highlighting the nature of a few prominent cysteine cathepsins, their involvement in select diseases, and the application of cathepsin imaging probes in research spanning basic biochemical studies to clinical applications.
Collapse
Affiliation(s)
- Kelton A Schleyer
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr, Gainesville, FL 32610, USA.
| | - Lina Cui
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr, Gainesville, FL 32610, USA.
| |
Collapse
|
38
|
Iyer SG. Fluorescence imaging in hepatobiliary surgery - hope and hype. Singapore Med J 2021; 62:157-158. [PMID: 33948666 DOI: 10.11622/smedj.2021035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Shridhar Ganpathi Iyer
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Singapore.,Liver Transplant Programme, National University Hospital, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
39
|
Sun R, Cuthbert H, Watts C. Fluorescence-Guided Surgery in the Surgical Treatment of Gliomas: Past, Present and Future. Cancers (Basel) 2021; 13:cancers13143508. [PMID: 34298721 PMCID: PMC8304525 DOI: 10.3390/cancers13143508] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Gliomas are aggressive central nervous system tumours. The emergence and recent widespread adoption of 5-aminolevulinic acid and fluorescence guided surgery have improved the extent of resection, with implications for improved survival and progression-free survival. This review describes the history, rationale and mechanism behind the use of 5-aminolevulinic acid and fluorescence-guided surgery. We also discuss current limitations and future directions for this important adjunct to glioma surgery. This review aims to provide readers with an up-to-date overview and evidence base on this important topic. Abstract Gliomas are central nervous systems tumours which are diffusely infiltrative and difficult to treat. The extent of surgical resection is correlated with improved outcomes, including survival and disease-free progression. Cancerous tissue can be directly visualised intra-operatively under fluorescence by administration of 5-aminolevulinic acid to the patient. The adoption of this technique has allowed surgeons worldwide to achieve greater extents of resection, with implications for improved prognosis. However, there are practical limitations to use of 5-aminolevulinic acid. New adjuncts in the field of fluorescence-guided surgery aim to improve recognition of the interface between tumour and brain with the objective of improving resection and patient outcomes.
Collapse
Affiliation(s)
- Rosa Sun
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham B15 2GW, UK;
- Correspondence: (R.S.); (H.C.)
| | - Hadleigh Cuthbert
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham B15 2GW, UK;
- Correspondence: (R.S.); (H.C.)
| | - Colin Watts
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham B15 2GW, UK;
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK
| |
Collapse
|
40
|
Simón M, Jørgensen JT, Juhl K, Kjaer A. The use of a uPAR-targeted probe for photothermal cancer therapy prolongs survival in a xenograft mouse model of glioblastoma. Oncotarget 2021; 12:1366-1376. [PMID: 34262647 PMCID: PMC8274719 DOI: 10.18632/oncotarget.28013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/14/2021] [Indexed: 01/10/2023] Open
Abstract
The development of tumor-targeted probes that can efficiently reach cancerous tissue is an important focus of preclinical research. Photothermal cancer therapy (PTT) relies on light-absorbing molecules, which are directed towards tumor tissue and irradiated with an external source of light. This light is transformed into heat, causing localized hyperthermia and tumor death. The fluorescent probe indocyanine green (ICG) is already used as an imaging agent both preclinically and in clinical settings, but its use for PTT is yet to be fully exploited due to its short retention time and non-specific tumor targeting. Therefore, increasing ICG tumor uptake is necessary to improve treatment outcome. The urokinase-type plasminogen activator receptor, uPAR, is overexpressed in multiple tumor types. ICG-Glu-Glu-AE105, consisting of the uPAR-targeting peptide AE105 conjugated to ICG, has shown great potential for fluorescence-guided surgery. In this study, ICG-Glu-Glu-AE105 was evaluated as photothermal agent in a subcutaneous mouse model of human glioblastoma. We observed that the photothermal abilities of ICG-Glu-Glu-AE105 triggered high temperatures in the tumor during PTT, leading to tumor death and prolonged survival. This confirms the potential of ICG-Glu-Glu-AE105 as photothermal agent and indicates that it could be used as an add-on to the application of the probe for fluorescence-guided surgery.
Collapse
Affiliation(s)
- Marina Simón
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Jesper Tranekjær Jørgensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Karina Juhl
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
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: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [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
|
42
|
Alfonso-Garcia A, Bec J, Weyers B, Marsden M, Zhou X, Li C, Marcu L. Mesoscopic fluorescence lifetime imaging: Fundamental principles, clinical applications and future directions. JOURNAL OF BIOPHOTONICS 2021; 14:e202000472. [PMID: 33710785 PMCID: PMC8579869 DOI: 10.1002/jbio.202000472] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 05/16/2023]
Abstract
Fluorescence lifetime imaging (FLIm) is an optical spectroscopic imaging technique capable of real-time assessments of tissue properties in clinical settings. Label-free FLIm is sensitive to changes in tissue structure and biochemistry resulting from pathological conditions, thus providing optical contrast to identify and monitor the progression of disease. Technical and methodological advances over the last two decades have enabled the development of FLIm instrumentation for real-time, in situ, mesoscopic imaging compatible with standard clinical workflows. Herein, we review the fundamental working principles of mesoscopic FLIm, discuss the technical characteristics of current clinical FLIm instrumentation, highlight the most commonly used analytical methods to interpret fluorescence lifetime data and discuss the recent applications of FLIm in surgical oncology and cardiovascular diagnostics. Finally, we conclude with an outlook on the future directions of clinical FLIm.
Collapse
Affiliation(s)
- Alba Alfonso-Garcia
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Julien Bec
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Brent Weyers
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Mark Marsden
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Xiangnan Zhou
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Cai Li
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Laura Marcu
- Department of Biomedical Engineering, University of California, Davis, Davis, California
- Department Neurological Surgery, University of California, Davis, California
| |
Collapse
|
43
|
Luciano MP, Dingle I, Nourian S, Schnermann MJ. Preferential Light-Chain Labeling of Native Monoclonal Antibodies Improves the Properties of Fluorophore Conjugates. Tetrahedron Lett 2021; 75. [PMID: 34321699 DOI: 10.1016/j.tetlet.2021.153211] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Site specific labeling methods have significant potential to enhance the properties of antibody conjugates. While studied extensively in the context of antibody-drug conjugates (ADCs), few studies have examined the impact of homogenous labeling on the properties of antibody-fluorophore conjugates (AFCs). We report the application of pentafluorophenyl (PFP) esters, which had previously been shown to be reasonably selective for K188 of the kappa light chain of human IGG antibodies, toward producing AFCs. We show that simple replacement of N-hydroxy succinimide (NHS) with PFP dramatically increases the light-chain specificity of near-infrared (NIR) AFCs. Comparing the properties of AFCs labeled using NHS and PFP-activated esters reveals that the latter exhibits reduced aggregation and improved brightness, both in vitro and in vivo. Overall, the use of PFP esters provides a remarkably simple approach to provide selectively labeled antibodies with improved properties.
Collapse
Affiliation(s)
- Michael P Luciano
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, United States
| | - Ivan Dingle
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, United States
| | - Saghar Nourian
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, United States
| |
Collapse
|
44
|
Usama SM, Inagaki F, Kobayashi H, Schnermann MJ. Norcyanine-Carbamates Are Versatile Near-Infrared Fluorogenic Probes. J Am Chem Soc 2021; 143:5674-5679. [PMID: 33844539 DOI: 10.1021/jacs.1c02112] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Fluorogenic probes in the near-infrared (NIR) region have the potential to provide stimuli-dependent information in living organisms. Here, we describe a new class of fluorogenic probes based on the heptamethine cyanine scaffold, the most broadly used NIR chromophore. These compounds result from modification of heptamethine norcyanines with stimuli-responsive carbamate linkers. The resulting cyanine carbamates (CyBams) exhibit exceptional turn-ON ratios (∼170×) due to dual requirements for NIR emission: carbamate cleavage through 1,6-elimination and chromophore protonation. Illustrating their utility in complex in vivo settings, a γ-glutamate substituted CyBam was applied to imaging γ-glutamyl transpeptidase (GGT) activity in a metastatic model of ovarian cancer. Overall, CyBams have significant potential to extend the reach of fluorogenic strategies to intact tissue and live animal imaging applications.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Fuyuki Inagaki
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
45
|
Anatase Titanium Dioxide Imparts Photoluminescent Properties to PA2200 Commercial 3D Printing Material to Generate Complex Optical Imaging Phantoms. MATERIALS 2021; 14:ma14071813. [PMID: 33917612 PMCID: PMC8038817 DOI: 10.3390/ma14071813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 01/06/2023]
Abstract
Selective laser sintering (SLS) is a prominent 3D printing modality that typically uses a polyamide (PA) powder as the substrate. One commercially available SLS material is known as PA2200, which is comprised of nylon 12 and titanium dioxide (TiO2) and is widely used to generate 3D-printed parts. Here, we report a unique optical photoluminescence (PL) characteristic of native, white PA2200, in which it yields a persistent, phosphorescence-type emission. An analysis of luminescence imaging data with emission measurements demonstrated that the anatase phase of the titanium dioxide additive is the source of the persistent PL properties. This characteristic of PA2200 enables advanced optical imaging applications, as demonstrated by luminescence imaging of an anatomical rat skeleton and a novel Derenzo-type phantom on a commercial image station. In summary, the light emission properties of PA2200 induced by the presence of anatase titanium dioxide open the door to a vast new array of complex optical applications, including the generation of imaging phantoms for training, calibration, and quality control.
Collapse
|
46
|
Tumor Accumulation and Off-Target Biodistribution of an Indocyanine-Green Fluorescent Nanotracer: An Ex Vivo Study on an Orthotopic Murine Model of Breast Cancer. Int J Mol Sci 2021; 22:ijms22041601. [PMID: 33562574 PMCID: PMC7915532 DOI: 10.3390/ijms22041601] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/15/2022] Open
Abstract
Indocyanine green (ICG) is a near infrared fluorescent tracer used in image-guided surgery to assist surgeons during resection. Despite appearing as a very promising tool for surgical oncology, its employment in this area is limited to lymph node mapping or to laparoscopic surgery, as it lacks tumor targeting specificity. Recently, a nanoformulation of this dye has been proposed with the aim toward tumor targeting specificity in order to expand its employment in surgical oncology. This nanosystem is constituted by 24 monomers of H-Ferritin (HFn), which self-assemble into a spherical cage structure enclosing the indocyanine green fluorescent tracer. These HFn nanocages were demonstrated to display tumor homing due to the specific interaction between the HFn nanocage and transferrin receptor 1, which is overexpressed in most tumor tissues. Here, we provide an ex vivo detailed comparison between the biodistribution of this nanotracer and free ICG, combining the results obtained with the Karl Storz endoscope that is currently used in clinical practice and the quantification of the ICG signal derived from the fluorescence imaging system IVIS Lumina II. These insights demonstrate the suitability of this novel HFn-based nanosystem in fluorescence-guided oncological surgery.
Collapse
|
47
|
Candidate Biomarkers for Specific Intraoperative Near-Infrared Imaging of Soft Tissue Sarcomas: A Systematic Review. Cancers (Basel) 2021; 13:cancers13030557. [PMID: 33535618 PMCID: PMC7867119 DOI: 10.3390/cancers13030557] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Near-infrared imaging of tumors during surgery facilitates the oncologic surgeon to distinguish malignant from healthy tissue. The technique is based on fluorescent tracers binding to tumor biomarkers on malignant cells. Currently, there are no clinically available fluorescent tracers that specifically target soft tissue sarcomas. This review searched the literature to find candidate biomarkers for soft tissue sarcomas, based on clinically used therapeutic antibodies. The search revealed 7 biomarkers: TEM1, VEGFR-1, EGFR, VEGFR-2, IGF-1R, PDGFRα, and CD40. These biomarkers are abundantly present on soft tissue sarcoma tumor cells and are already being targeted with humanized monoclonal antibodies. The conjugation of these antibodies with a fluorescent dye will yield in specific tracers for image-guided surgery of soft tissue sarcomas to improve the success rates of tumor resections. Abstract Surgery is the mainstay of treatment for localized soft tissue sarcomas (STS). The curative treatment highly depends on complete tumor resection, as positive margins are associated with local recurrence (LR) and prognosis. However, determining the tumor margin during surgery is challenging. Real-time tumor-specific imaging can facilitate complete resection by visualizing tumor tissue during surgery. Unfortunately, STS specific tracers are presently not clinically available. In this review, STS-associated cell surface-expressed biomarkers, which are currently already clinically targeted with monoclonal antibodies for therapeutic purposes, are evaluated for their use in near-infrared fluorescence (NIRF) imaging of STS. Clinically targeted biomarkers in STS were extracted from clinical trial registers and a PubMed search was performed. Data on biomarker characteristics, sample size, percentage of biomarker-positive STS samples, pattern of biomarker expression, biomarker internalization features, and previous applications of the biomarker in imaging were extracted. The biomarkers were ranked utilizing a previously described scoring system. Eleven cell surface-expressed biomarkers were identified from which 7 were selected as potential biomarkers for NIRF imaging: TEM1, VEGFR-1, EGFR, VEGFR-2, IGF-1R, PDGFRα, and CD40. Promising biomarkers in common and aggressive STS subtypes are TEM1 for myxofibrosarcoma, TEM1, and PDGFRα for undifferentiated soft tissue sarcoma and EGFR for synovial sarcoma.
Collapse
|
48
|
Daoust F, Nguyen T, Orsini P, Bismuth J, de Denus-Baillargeon MM, Veilleux I, Wetter A, Mckoy P, Dicaire I, Massabki M, Petrecca K, Leblond F. Handheld macroscopic Raman spectroscopy imaging instrument for machine-learning-based molecular tissue margins characterization. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-200306SSR. [PMID: 33580641 PMCID: PMC7880244 DOI: 10.1117/1.jbo.26.2.022911] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/19/2021] [Indexed: 05/08/2023]
Abstract
SIGNIFICANCE Raman spectroscopy has been developed for surgical guidance applications interrogating live tissue during tumor resection procedures to detect molecular contrast consistent with cancer pathophysiological changes. To date, the vibrational spectroscopy systems developed for medical applications include single-point measurement probes and intraoperative microscopes. There is a need to develop systems with larger fields of view (FOVs) for rapid intraoperative cancer margin detection during surgery. AIM We design a handheld macroscopic Raman imaging system for in vivo tissue margin characterization and test its performance in a model system. APPROACH The system is made of a sterilizable line scanner employing a coherent fiber bundle for relaying excitation light from a 785-nm laser to the tissue. A second coherent fiber bundle is used for hyperspectral detection of the fingerprint Raman signal over an area of 1 cm2. Machine learning classifiers were trained and validated on porcine adipose and muscle tissue. RESULTS Porcine adipose versus muscle margin detection was validated ex vivo with an accuracy of 99% over the FOV of 95 mm2 in ∼3 min using a support vector machine. CONCLUSIONS This system is the first large FOV Raman imaging system designed to be integrated in the workflow of surgical cancer resection. It will be further improved with the aim of discriminating brain cancer in a clinically acceptable timeframe during glioma surgery.
Collapse
Affiliation(s)
- François Daoust
- Polytechnique Montreal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Tien Nguyen
- Polytechnique Montreal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | - Israel Veilleux
- Polytechnique Montreal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | - Kevin Petrecca
- McGill University, Montreal Neurological Institute-Hospital, Department of Neurology and Neurosurgery, Montreal, Quebec, Canada
| | - Frédéric Leblond
- Polytechnique Montreal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Address all correspondence to Frédéric Leblond,
| |
Collapse
|
49
|
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: 2.3] [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
|
50
|
Montecinos-Franjola F, Lin JY, Rodriguez EA. Fluorescent proteins for in vivo imaging, where's the biliverdin? Biochem Soc Trans 2020; 48:2657-2667. [PMID: 33196077 DOI: 10.1042/bst20200444] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022]
Abstract
Noninvasive fluorescent imaging requires far-red and near-infrared fluorescent proteins for deeper imaging. Near-infrared light penetrates biological tissue with blood vessels due to low absorbance, scattering, and reflection of light and has a greater signal-to-noise due to less autofluorescence. Far-red and near-infrared fluorescent proteins absorb light >600 nm to expand the color palette for imaging multiple biosensors and noninvasive in vivo imaging. The ideal fluorescent proteins are bright, photobleach minimally, express well in the desired cells, do not oligomerize, and generate or incorporate exogenous fluorophores efficiently. Coral-derived red fluorescent proteins require oxygen for fluorophore formation and release two hydrogen peroxide molecules. New fluorescent proteins based on phytochrome and phycobiliproteins use biliverdin IXα as fluorophores, do not require oxygen for maturation to image anaerobic organisms and tumor core, and do not generate hydrogen peroxide. The small Ultra-Red Fluorescent Protein (smURFP) was evolved from a cyanobacterial phycobiliprotein to covalently attach biliverdin as an exogenous fluorophore. The small Ultra-Red Fluorescent Protein is biophysically as bright as the enhanced green fluorescent protein, is exceptionally photostable, used for biosensor development, and visible in living mice. Novel applications of smURFP include in vitro protein diagnostics with attomolar (10-18 M) sensitivity, encapsulation in viral particles, and fluorescent protein nanoparticles. However, the availability of biliverdin limits the fluorescence of biliverdin-attaching fluorescent proteins; hence, extra biliverdin is needed to enhance brightness. New methods for improved biliverdin bioavailability are necessary to develop improved bright far-red and near-infrared fluorescent proteins for noninvasive imaging in vivo.
Collapse
Affiliation(s)
| | - John Y Lin
- School of Medicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Erik A Rodriguez
- Department of Chemistry, The George Washington University, Washington, DC 20052, U.S.A
| |
Collapse
|