1
|
Wang Z, Dai B, Li Y, Cao Y, Wang D, Liu F, Li Z, Cai H, Butch CJ, Wang Y, Nie S. Signal-to-Noise Ratio Imaging and Real-Time Sharpening of Tumor Boundaries for Image-Guided Cancer Surgery. Anal Chem 2025; 97:8516-8527. [PMID: 40193701 DOI: 10.1021/acs.analchem.5c00530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Fluorescence-guided cancer surgery is of considerable current interest in bioanalytical chemistry, engineering, and medicine, but its clinical utility is still hampered by the diffusive (scattering) nature of human tissues and large variations among different patients. Here, we report a new method based on signal-to-noise (contrast-to-noise) ratio (SNR or CNR) imaging for real-time delineation and sharpening of tumor boundaries during image-guided cancer surgery. In particular, we show that in vivo tumor fluorescence signals (both intensity and standard deviation) are strongly correlated with those of the surrounding tissue of the same tissue type and that this relationship is maintained as a function of time for fluorescent tracers such as indocyanine green. This dynamic relationship permits a precise removal of nonspecific background fluorescence from tumor fluorescence. As a result, single-pixel SNR values have been calculated, mapped, and displayed across a large surgical field at 60 frames per second. Pathological validation studies indicate that these SNR values correspond to statistical confidence levels similar (but not identical) to those of normal distributions. When the tumor fluorescence has an SNR of 3, pathological data show a confidence level of approximately 95% in identifying the true tumor lesions. For clinical relevance, we have also carried out first-in-human clinical studies for both oral and esophageal tumors, achieving tumor margin precisions of 1-2 mm with 87.5% histological accuracy and no false positives.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Bo Dai
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing 210008, China
| | - Yunlong Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Ying Cao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Dong Wang
- Department of Thoracic Surgery, Taikang Xianlin Drum Tower Hospital, Nanjing University School of Medicine, Nanjing 210008, China
| | - Fayu Liu
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, Liaoning Province Key Laboratory of Oral Disease, China Medical University, Shenyang 110052, China
| | - Zhenning Li
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, Liaoning Province Key Laboratory of Oral Disease, China Medical University, Shenyang 110052, China
| | - Huiming Cai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
- Nanjing Nuoyuan Medical Devices Co. Ltd, Nanjing 211514, China
| | - Christopher J Butch
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Yiqing Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China
| | - Shuming Nie
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana 61801, United States
| |
Collapse
|
2
|
Umadevi K, Sundeep D, Vighnesh AR, Misra A, Krishna AG. Current Trends and Advances in Nanoplatforms-Based Imaging for Cancer Diagnosis. Indian J Microbiol 2025; 65:137-176. [PMID: 40371042 PMCID: PMC12069201 DOI: 10.1007/s12088-024-01373-9] [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: 05/05/2024] [Accepted: 08/02/2024] [Indexed: 05/16/2025] Open
Abstract
The intersection of nanotechnology and biomedical imaging has ushered in a new era in the early detection and diagnosis of cancer which has revolutionized biomedical imaging by enhancing sensitivity, resolution, and targeting capability. This review presents a comprehensive overview of the latest developments and innovations in nanoplatforms-based imaging for cancer diagnosis, a burgeoning field that holds significant potential in improving cancer detection and treatment. Recently multimodal imaging techniques utilizing the unique properties of different types of nanoparticles are providing comprehensive diagnostic information. This multi-pronged approach allows for more precise tumor localization, size estimation, and growth rate calculation, offering a holistic view of the tumor and its environment. The primary focus of this review is on the recent progress in various types of nanoparticle-based imaging modalities, including optical, magnetic resonance, ultrasound, and nuclear imaging. Specific advancements in nanomaterial design for targeted imaging are highlighted, showing the improvement of precision targeting as an impact on the detection of cancer cells, even in early-stage tumors. A keen examination on the integration of diagnostic and therapeutic capabilities into single nano-based platforms for theranostics, underscoring their potential in personalized medicine is provided. The current challenges in the field, such as issues related to toxicity, biodistribution, and clearance of nanoparticles, and it explores ongoing research aimed at overcoming these hurdles. The growing body of research in this field highlights the promising future of nanoplatforms in improving the early detection and treatment of cancer.
Collapse
Affiliation(s)
- Kovuri Umadevi
- Department of Pathology, Government Medical College and Hospital, Khaleelwadi, Nizamabad, Telangana 503001 India
| | - Dola Sundeep
- Biomedical Research Laboratory, Department of Electronics and Communication Engineering, Indian Institute of Information Technology Design and Manufacturing, Jagannathagattu Hill, Kurnool, Andhra Pradesh 518008 India
| | - Alluru Raghavendra Vighnesh
- Department of Mechanical Engineering, Indian Institute of Technology (IIT-BHU) Varanasi, Varanasi, Uttar Pradesh 221005 India
| | - Aroonima Misra
- ICMR-National Institute of Pathology, Sadarjang Hospital Campus, Ansari Nagar West, New Delhi, Delhi 110029 India
| | - Alluru Gopala Krishna
- Department of Mechanical Engineering, Jawaharlal Nehru Technological University Kakinada, Nagamallithota, Kakinada, Andhra Pradesh 533003 India
| |
Collapse
|
3
|
Yang T, Zhang Q, Miao Y, Lyu Y, Xu Y, Yang M, Mao C. Tumor-Homing Phage Nanofibers for Nanozyme-Enhanced Targeted Breast Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2403756. [PMID: 39233557 PMCID: PMC11733710 DOI: 10.1002/adma.202403756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/24/2024] [Indexed: 09/06/2024]
Abstract
Photodynamic therapy (PDT) eliminates cancer cells by converting endogenous oxygen into reactive oxygen species (ROS). However, its efficacy is significantly hindered by hypoxia in solid tumors. Hence, to engineer filamentous fd phage, a human-friendly bacteria-specific virus is proposed, into a nanozyme-nucleating photosensitizer-loaded tumor-homing nanofiber for enhanced production of ROS in a hypoxic tumor. Specifically, Pt-binding and tumor-homing peptides are genetically displayed on the sidewall and tip of the fd phage, respectively. The Pt-binding peptides induced nucleation and orientation of Pt nanozymes (PtNEs) on the sidewall of the phage. The resultant PtNE-coated tumor-homing phage exhibits significantly enhanced sustained catalytic conversion of hydrogen peroxide in hypoxic tumors into O2 for producing ROS needed for PDT, compared to non-phage-templated PtNE. Density functional theory (DFT) calculations verify the catalytic mechanism of the phage-templated PtNE. After intravenous injection of the PtNE-coated indocyanine green (ICG)-loaded tumor-homing phages into breast tumor-bearing mice, the nanofibers home to the tumors and effectively inhibit tumor growth by the PtNE-enhanced PDT. The nanofibers can also serve as a tumor-homing imaging probe due to the fluorescence of ICG. This work demonstrates that filamentous phage, engineered to become tumor-homing nanozyme-nucleating tumor-hypoxia-relieving nanofibers, can act as cancer-targeting nanozymes with improved catalytic performance for effective targeted PDT.
Collapse
Affiliation(s)
- Tao Yang
- School of Materials Science & EngineeringZhejiang UniversityHangzhouZhejiang310027P. R. China
- Department of Biomedical EngineeringThe Chinese University of Hong KongSha TinHong Kong SARP. R. China
| | - Qinglei Zhang
- Institute of Applied Bioresource ResearchCollege of Animal ScienceZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Yao Miao
- School of Materials Science & EngineeringZhejiang UniversityHangzhouZhejiang310027P. R. China
| | - Yang Lyu
- Department of Biomedical EngineeringThe Chinese University of Hong KongSha TinHong Kong SARP. R. China
| | - Yajing Xu
- Department of Biomedical EngineeringThe Chinese University of Hong KongSha TinHong Kong SARP. R. China
| | - Mingying Yang
- Institute of Applied Bioresource ResearchCollege of Animal ScienceZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Chuanbin Mao
- Department of Biomedical EngineeringThe Chinese University of Hong KongSha TinHong Kong SARP. R. China
| |
Collapse
|
4
|
Wongso H, Kurniawan A, Setiadi Y, Kusumaningrum CE, Widyasari EM, Wibawa TH, Mahendra I, Febrian MB, Sriyani ME, Halimah I, Daruwati I, Gunawan R, Achmad A, Nugraha DH, Lesmana R, Nugraha AS. Translocator Protein 18 kDa (TSPO): A Promising Molecular Target for Image-Guided Surgery of Solid Cancers. Adv Pharm Bull 2024; 14:86-104. [PMID: 38585455 PMCID: PMC10997928 DOI: 10.34172/apb.2024.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/26/2023] [Accepted: 10/08/2023] [Indexed: 04/09/2024] Open
Abstract
The translocator protein 18-kDa (TSPO) is a mitochondrial membrane protein that is previously identified as the peripheral benzodiazepine receptor (PBR). Furthermore, it plays a significant role in a diverse range of biochemical processes, including steroidogenesis, mitochondrial cholesterol transport, cell survival and death, cell proliferation, and carcinogenesis. Several investigations also reported its roles in various types of cancers, including colorectal, brain, breast, prostate, and lung cancers, as well as melanoma. According to a previous study, the expression of TSPO was upregulated in cancer cells, which corresponds to an aggressive phenotype and/or poor prognosis. Consequently, the potential for crafting diagnostic and prognostic tools with a focus on TSPO holds great potential. In this context, several radioligands designed to target this protein have been identified, and some of the candidates have advanced to clinical trials. In recent years, the use of hybrid probes with radioactive and fluorescence molecules for image-guided surgery has exhibited promising results in animal and human studies. This indicates that the approach can serve as a valuable surgical navigator during cancer surgery. The current hybrid probes are built from various molecular platforms, including small molecules, nanoparticles, and antibodies. Although several TSPO-targeted imaging probes have been developed, their development for image-guided surgery of cancers is still limited. Therefore, this review aims to highlight recent findings on the involvement of TSPO in carcinogenesis, as well as provide a new perspective on the potential application of TSPO-targeted hybrid probes for image-guided surgery.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Ahmad Kurniawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Yanuar Setiadi
- Research Center for Environmental and Clean Technology, Research Organization for Life Sciences and Environment, National Research and Innovation Agency, Puspiptek, Banten 15314, Indonesia
| | - Crhisterra E. Kusumaningrum
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Eva M. Widyasari
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Teguh H.A. Wibawa
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Isa Mahendra
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Muhamad B. Febrian
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Maula E. Sriyani
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Iim Halimah
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
| | - Isti Daruwati
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Rudi Gunawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency Republic of Indonesia, Puspiptek, Banten 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
| | - Arifudin Achmad
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Ir. Soekarno KM 21, Jatinangor 45363, Indonesia
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161
- Oncology and Stem Cells Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161
| | | | - Ronny Lesmana
- Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Physiology Molecular, Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Ari S. Nugraha
- Drug Utilisation and Discovery Research Group, Faculty of Pharmacy, Universitas Jember, Jember 68121, Indonesia
- School of Chemistry and Molecular Biosciences, Molecular Horizons, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| |
Collapse
|
5
|
Akopov AL, Papayan GV, Fedotova DA. [Intraoperative near-infrared fluorescence imaging of peripheral lung tumors]. Khirurgiia (Mosk) 2024:79-85. [PMID: 38258692 DOI: 10.17116/hirurgia202401179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The main objective of surgical intervention in lung cancer is the radical removal of the entire tumor with the maximum possible preservation of healthy tissue. Although the area of the tumor lesion is known by the results of preoperative studies, it can be difficult to use this information to establish the exact boundaries of resection during surgery, especially with small sizes of the lump and when using minimally invasive approaches. There are several techniques to solve this problem. One of the latter is intraoperative fluorescence imaging in the infrared range, which makes it possible to detect a tumor not only with greater contrast than it can be done in white light, but also with its deep location. This review is devoted to the discussion of various aspects of this approach related to molecular imaging methods. The current situation based on the use of green indocyanine green, available for clinical use as a fluorescent agent is considered, the issues of using new targeted drugs are examined, as well as the possibility of increasing the depth of probing and combining with related treatment methods, which should contribute to a more radical operation and reduce the likelihood of local relapses.
Collapse
Affiliation(s)
- A L Akopov
- Pavlov First State Medical University of St. Petersburg, St. Petersburg, Russia
| | - G V Papayan
- Pavlov First State Medical University of St. Petersburg, St. Petersburg, Russia
- V.A. Almazov NMRC, St. Petersburg, Russia
| | - D A Fedotova
- Pavlov First State Medical University of St. Petersburg, St. Petersburg, Russia
| |
Collapse
|
6
|
Wongso H, Kurniawan A, Forentin AM, Susilo VY, Setiadi Y, Mahendra I, Febrian MB, Rosdianto AM, Setiawan I, Goenawan H, Susianti S, Supratman U, Widyasari EM, Wibawa TH, Sriyani ME, Halimah I, Lesmana R. New hybrid radio-fluorescent probes [ 131I]-BPF-01 and [ 131I]-BPF-02 for visualisation of cancer cells: Synthesis and preliminary in vitro and ex vivo evaluations. Heliyon 2023; 9:e20710. [PMID: 37860547 PMCID: PMC10582398 DOI: 10.1016/j.heliyon.2023.e20710] [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: 04/30/2023] [Revised: 09/30/2023] [Accepted: 10/04/2023] [Indexed: 10/21/2023] Open
Abstract
We synthesised and biologically evaluated two new hybrid probes [131I]BPF-01 and [131I]BPF-02 which were built from three structural entities: benzothiazole-phenyl, fluorescein isothiocyanate (FITC), and iodine-131. These probes were designed for potential applications in assisting surgical procedures of solid cancers. The cytotoxicity study demonstrated that fluorescent probes BPF-01 (31.23 μg/mL) and BPF-02 (250 μg/mL) were relatively not toxic to normal immortalized human keratinocytes (HaCaT) cells, as indicated by the percentage of cell survival above 50 %. Furthermore, both probes displayed low to moderate anticancer activity against the breast cancer cells (MDA-MB-231) and prostate cancer cells (LNCaP and DU-145). The probe BPF-01 apparently showed an accumulation in the tumour tissues, as suggested by ex vivo fluorescence examinations. In addition, the cellular uptake study suggests that hybrid probe [131I]-BPF-01 was potentially accumulated in the MCF-7 cell line with the highest uptake of 16.11 ± 1.52 % after 2 h of incubation, approximately 50-fold higher than the accumulation of iodine-131 (control). The magnetic bead assay suggests that [131I]-BPF-02 and [131I]-BPF-02 showed a promising capability to interact with translocator protein 18 kDa (TSPO). Moreover, the computational data showed that the binding scores for ligands 7-8, BPF-01 and BPF-02, and [131I]-BPF-01 and [131I]-BPF-02 in the TSPO were considerably high. Accordingly, fluorescent probes BPF-01 and BPF-02, and hybrid probes [131I]BPF-01 and [131I]BPF-02 can be further developed for targeting cancer cells during intraoperative tumour surgery.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia
| | - Ahmad Kurniawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Alfian M. Forentin
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Veronika Y. Susilo
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Yanuar Setiadi
- Research Center for Environmental and Clean Technology, Research Organization for Life Sciences and Environment, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Isa Mahendra
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia
| | - Muhamad B. Febrian
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Aziiz M. Rosdianto
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Iwan Setiawan
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Hanna Goenawan
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Susianti Susianti
- Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Unang Supratman
- Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Eva M. Widyasari
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Teguh H.A. Wibawa
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Maula E. Sriyani
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Iim Halimah
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Ronny Lesmana
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
7
|
Wongso H, Goenawan H, Lesmana R, Mahendra I, Kurniawan A, Wibawa THA, Nuraeni W, Rosyidiah E, Setiadi Y, Sylviana N, Pratiwi YS, Rosdianto AM, Supratman U, Kusumaningrum CE. Synthesis and Biological Evaluation of New Fluorescent Probe BPN-01: A Model Molecule for Fluorescence Image-guided Surgery. J Fluoresc 2023; 33:1827-1839. [PMID: 36847931 DOI: 10.1007/s10895-023-03166-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/02/2023] [Indexed: 03/01/2023]
Abstract
Fluorescence image-guided surgery (FIGS) can serve as a tool to achieve successful resection of tumour tissues during surgery, serving as a surgical navigator for surgeons. FIGS relies on the use of fluorescent molecules that can specifically interact with cancer cells. In this work, we developed a new model of fluorescent probe based on benzothiazole-phenylamide moiety featuring the visible fluorophore nitrobenzoxadiazole (NBD), namely BPN-01. This compound was designed and synthesised for potential applications in the tissue biopsy examination and ex-vivo imaging during FIGS of solid cancers. The probe BPN-01 exhibited favourable spectroscopic properties, particularly in nonpolar and alkaline solvents. Moreover, in vitro fluorescence imaging revealed that the probe appeared to recognise and be internalised in the prostate (DU-145) and melanoma (B16-F10) cancer cells, but not in the normal cells (myoblast C2C12). The cytotoxicity studies revealed that probe BPN-01 was not toxic to the B16 cells, suggesting excellent biocompatibility. Furthermore, the computational analysis showed that the calculated binding affinity of the probe to both translocator protein 18 kDa (TSPO) and human epidermal growth factor receptor 2 (HER2) was considerably high. Hence, probe BPN-01 displays promising properties and may be valuable for visualising cancer cells in vitro. Furthermore, ligand 5 can potentially be labelled with NIR fluorophore and radionuclide, and serves as a dual imaging agent for in vivo applications.
Collapse
Affiliation(s)
- Hendris Wongso
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia.
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia.
| | - Hanna Goenawan
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Physiology Molecular, Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Ronny Lesmana
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Physiology Molecular, Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Isa Mahendra
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
- Research Collaboration Center for Theranostic Radiopharmaceuticals, National Research and Innovation Agency, Jl. Raya Bandung-Sumedang KM 21, Sumedang, 45363, Indonesia
| | - Ahmad Kurniawan
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Teguh H A Wibawa
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Witri Nuraeni
- Directorate of Laboratory Management, Research Facilities, and Science and Technology Park, National Research and Innovation Agency of Indonesia, Jl. Tamansari No. 71, Lb. Siliwangi, Bandung, West Java, 40132, Indonesia
| | - Endah Rosyidiah
- Directorate of Laboratory Management, Research Facilities, and Science and Technology Park, National Research and Innovation Agency of Indonesia, Jl. Tamansari No. 71, Lb. Siliwangi, Bandung, West Java, 40132, Indonesia
| | - Yanuar Setiadi
- Research Organization for Life Sciences and Environment, Research Center for Environmental and Clean Technology, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| | - Nova Sylviana
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Physiology Molecular, Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Yuni Susanti Pratiwi
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Physiology Molecular, Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| | - Aziiz Mardanarian Rosdianto
- Department of Biomedical Science, Physiology Division, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Physiology Molecular, Division of Biological Activity, Central Laboratory, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
- Laboratory of Sciences, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
- Veterinary Medicine Study Program, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, 45363, Indonesia
| | - Unang Supratman
- Departement of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km 21, Jatinangor, 45363, Indonesia
| | - Crhisterra E Kusumaningrum
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, Research Organization for Nuclear Energy, National Research and Innovation Agency, Puspiptek, Banten, 15314, Indonesia
| |
Collapse
|
8
|
Kumar BS. Desorption electrospray ionization mass spectrometry imaging (DESI-MSI) in disease diagnosis: an overview. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:3768-3784. [PMID: 37503728 DOI: 10.1039/d3ay00867c] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Tissue analysis, which is essential to histology and is considered the benchmark for the diagnosis and prognosis of many illnesses, including cancer, is significant. During surgery, the surgical margin of the tumor is assessed using the labor-intensive, challenging, and commonly subjective technique known as frozen section histopathology. In the biopsy section, large numbers of molecules can now be visualized at once (ion images) following recent developments in [MSI] mass spectrometry imaging under atmospheric conditions. This is vastly superior to and different from the single optical tissue image processing used in traditional histopathology. This review article will focus on the advancement of desorption electrospray ionization mass spectrometry imaging [DESI-MSI] technique, which is label-free and requires little to no sample preparation. Since the proportion of molecular species in normal and abnormal tissues is different, DESI-MSI can capture ion images of the distributions of lipids and metabolites on biopsy sections, which can provide rich diagnostic information. This is not a systematic review but a summary of well-known, cutting-edge and recent DESI-MSI applications in cancer research between 2018 and 2023.
Collapse
Affiliation(s)
- Bharath Sampath Kumar
- Independent Researcher, 21, B2, 27th Street, Nanganallur, Chennai 61, TamilNadu, India.
| |
Collapse
|
9
|
Hettie KS, Chin FT. NIRDye 812: A molecular platform tailored for multimodal bioimaging applications of targeted fluorescence- and photoacoustic-guided surgery. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 242:112683. [PMID: 36934549 DOI: 10.1016/j.jphotobiol.2023.112683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/16/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
The primary treatment for malignant tumors remains to be surgical removal of the diseased tissue. The presence or absence of residual diseased tissue at the tumor margin is the strongest predictor of postoperative prognosis and recurrence. Accordingly, reliance on the ability of surgeons to visually distinguish diseased tissue from healthy tissue unambiguously in real time is crucial. Near infrared-I (NIRI) fluorescence-emitting targeting biomolecular constructs such as anticancer antibody-fluorophore conjugates, namely cetuximab-IRDye® 800CW (CTB-IRDye® 800CW), are FDA-approved for clinical trial usage in the fluorescence-guided resection of diseased tissue due to affording improved direct visualization of tumor tissue when compared to the use of either the unaided eye under standard white light illumination (WLI) surgical techniques or non-targeting fluorophores. Unfortunately, though helpful, CTB-IRDye® 800CW affords limited (i) identification of diseased tissue and (ii) tumor margin delineation, because the immunoconjugate generates suboptimal tumor-to-background ratios (TBRs) as a result of its spectral/photophysical profiles poorly aligning with the fixed optical windows of pre-/clinical setups. As such, CTB-IRDye® 800CW is more prone to affording incomplete resection compared to if TBRs were higher due to otherwise. To aid in accurately identifying deep-seated diseased tissue, photoacoustic (PA) tomography has been implemented alongside CTB-IRDye® 800CW to achieve PA signals that could result in higher TBRs. However, in clinical trial practice, using IRDye® 800CW for PA imaging also yields subpar TBRs due to it affording low PA signals. To overcome such limitations, we developed NIRDye 812, a structurally-modified topological equivalent of IRDye® 800CW, to confer it the capability to yield both higher TBRs and superior PA signal than that of the equivalent CTB-conjugate and fluorophore IRDye® 800CW itself, respectively. To do so, we substituted the oxygen atom at its meso-position with a sulfur atom. CTB-NIRDye 812 demonstrated a red-shifted absorption wavelength at 796 nm and a peak NIR-I fluorescence emission wavelength at 820 nm, which better dovetails with the fixed windows of preinstalled fixed emission filters within commercial pre-/clinical NIR-I fluorescence imaging instruments. Overall, CTB-NIRDye 812 provided a ∼ 2-fold increase in TBRs compared to those of CTB-IRDye® 800CW in vivo. Also, NIRDye 812 displayed an ∼60% higher PA signal than that of IRDye® 800CW. Collectively, we achieved our goal of improving upon the spectral/photophysical and PA properties of IRDye® 800CW via introducing a subtle modification to its electronic core such that its CTB immunoconjugate could potentially allow for fast track or breakthrough designation by the FDA due to its near-identical structure displaying considerably improved efficacy.
Collapse
Affiliation(s)
- Kenneth S Hettie
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA; Department of Otolaryngology - Head & Neck Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Frederick T Chin
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
10
|
Quan YH, Oh CH, Jung D, Lim JY, Choi BH, Rho J, Choi Y, Han KN, Kim BM, Kim C, Park JH, Kim HK. Evaluation of Intraoperative Near-Infrared Fluorescence Visualization of the Lung Tumor Margin With Indocyanine Green Inhalation. JAMA Surg 2021; 155:732-740. [PMID: 32579150 DOI: 10.1001/jamasurg.2020.1314] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Importance Identification of the tumor margin during surgery is important for precise minimal resection of lung tumors. Intravenous injection of indocyanine green (ICG) has several limitations when used for intraoperative visualization of lung cancer. Objectives To describe a technique for intraoperative visualization of lung tumor margin using ICG inhalation and evaluate the clinical applicability of the technique in mouse and rabbit lung tumor models as well as lung specimens of patients with lung tumors. Design, Setting, and Participants In lung tumor models of both mice and rabbits, the distribution of inhaled ICG in the lung tumor margin was investigated in vivo and ex vivo using a near-infrared imaging system. Lung tumor margin detection via inhalation of ICG was evaluated by comparing the results obtained with those of the intravenous injection method (n = 32, each time point for 4 mice). Based on preclinical data, use of ICG inhalation to help detect the tumor margin in patients with lung cancer was also evaluated (n = 6). This diagnostic study was conducted from May 31, 2017, to March 30, 2019. Main Outcomes and Measures The use of tumor margin detection by inhaled ICG was evaluated by comparing the inhaled formulation with intravenous administration of ICG. Results From 10 minutes after inhalation of ICG to 24 hours, the distribution of ICG in the lungs was significantly higher than that in other organs (signal to noise ratio in the lungs: 39 486.4; interquartile range [IQR], 36 983.74-43 592.5). Ex vivo and histologic analysis showed that, in both lung tumor models, inhaled ICG was observed throughout the healthy lung tissue but was rarely found in tumor tissue. The difference in the fluorescent signal between healthy and tumor lung tissues was associated with the mechanical airway obstruction caused by the tumor and with alveolar macrophage uptake of the inhaled ICG in healthy tissues. Inhalation at a 20-fold lower dose of ICG had a 2-fold higher efficiency for tumor margin detection than did the intravenous injection (2.9; IQR, 2.7-3.2; P < .001). Conclusions and Relevance The results of this study suggest that lung-specific inhalation delivery of ICG is feasible and may be useful for the intraoperative visualization of lung tumor margin in clinical practice.
Collapse
Affiliation(s)
- Yu Hua Quan
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Chan Hee Oh
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Daeho Jung
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ji-Young Lim
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Byeong Hyeon Choi
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jiyun Rho
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yeonho Choi
- Department of Bio-Convergence, Korea University, Seoul, Republic of Korea
| | - Kook Nam Han
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Beop-Min Kim
- Department of Bio-Convergence, Korea University, Seoul, Republic of Korea
| | - Chungyeul Kim
- Department of Pathology, Korea University Guro Hospital, College of Medicine, Republic of Korea University, Seoul, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, College of Medicine, Korea University, Seoul, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| |
Collapse
|
11
|
Stevens RM, Silverman A, Pascasio J, Sun J, Throckmorton A, Mulinari L, Cusimano RJ. Multi-disciplinary frameworks for the treatment of primary cardiac sarcoma. J Card Surg 2021; 36:3456-3457. [PMID: 34131956 DOI: 10.1111/jocs.15742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Randy M Stevens
- Division of Pediatric Cardiac Surgery, Pediatric Cardiac Surgery, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Ayaka Silverman
- Department of Pathology, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Judy Pascasio
- Department of Pathology, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Jie Sun
- Division of Pediatric Cardiology, Pediatric Cardiology, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Amy Throckmorton
- BioCirc Research Laboratory, School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Leonardo Mulinari
- Pediatric and Congenital Cardiac Surgery, DeWitt Daughtry Family Department of Surgery, Congenital Heart Center, Holtz Children's Hospital, University of Miami Leonard Miller School of Medicine, Miami, Florida, USA
| | - Robert James Cusimano
- Division of Cardiac Surgery, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Protic M, Krsmanovic O, Solajic N, Kukic B, Nikolic I, Bogdanovic B, Radovanovic Z, Kresoja M, Mannion C, Man YG, Stojadinovic A. Prospective Non-Randomized Study of Intraoperative Assessment of Surgical Resection Margin of Colo-Rectal Liver Metastases. J Cancer 2021; 12:3701-3714. [PMID: 33995645 PMCID: PMC8120181 DOI: 10.7150/jca.58580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/19/2021] [Indexed: 01/10/2023] Open
Abstract
Introduction: More than 50% of patients with colorectal cancer (CRC) develop liver metastases during the natural course of disease. Surgical resection is currently the most potentially curative method in the treatment of colorectal liver metastases (CRLM). The goal of surgery is to achieve a negative resection margin (RM) of at least 1 mm, which provides the best prognosis for patients. The RM can be assessed by the pathologist of the resected liver specimen (RLS) and by the surgeon intraoperatively. The aim of this research paper is to determine the degree of agreement on intraoperative assessment of the RM by the surgeon and histopathological RM assessment by the pathologist. Material and methods: This prospective non-randomized double-blind study was approved by the Ethics Committee of the Oncology Institute of Vojvodina and registered on ClinicalTrials.gov #NCT04634526. The study was conducted at the Oncology Institute of Vojvodina, Sremska Kamenica, Serbia. An experienced hepatobiliary surgeon assessed RM for every specimen intra-operatively, immediately after CRLM resection. Resected CRLM lesions were analyzed by two experienced pathologists. These data were compared with pathological RM assessment as a “gold standard”. RM of 1 mm or more was rated as negative RM (RM-). Disease-free survival (DFS) and recurrence rate was calculated by RM status defined by surgeon and by pathologist. Results: From 01 January 2015 to 31 August 2019, 98 patients were enrolled in the study. There were 219 RLS with 245 CRLM. The surgeon registered positive RM (RM+) of <1mm in 41 (18.7%) RLS. Taking the result of the histopathological assessment (HPA) as the “gold standard”, it was determined that RM was true positive in 32 (14.6%) cases. False positive RM was found in 9 (4.1%) cases. False negative RM was found in 20 (9.1%) cases. True negative RM was found in 158 (72.2%) cases. Sensitivity of surgical assessment (SA) of RM+ was 61.5% (32/52). Specificity of SA of RM+ was 94.6% (158/167). The positive predictive value (PPV) was 78.0% (32/41), while the negative predictive value (NPV) was 88.8% (158/178). The overall accuracy of the RM+ SA was 86.8% (190/219). There was no statistically significant difference in the assessment of RM+ per RLS by surgeon and pathologists (p=0.061), but it was significant when analyses per patients was performed (p=0.017). Recurrence rate for RM+ patients was 48.1% (13/27, p=0.05) for SA and 35.0% (14/40, p=0.17) for HPA. Three year DFS for RM- and RM+ was 66.5% and 27.9% (p=0.04), respectively, by SA, and 64.8% and 42.1% (p=0.106), respectively, by HPA. Conclusion: Intraoperative assessment of RM- by surgeon of RLS is clinically meaningful. There is not a statistically significant difference in the assessment of RM+ by surgeon and pathologists per RLS, but it was statically significant on a per patient basis. RM determined by surgeon has better prognostic impact on recurrence rate and 1- and 3-year DFS than standard histopathological assessment.
Collapse
Affiliation(s)
- Mladjan Protic
- Clinic for Surgical Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, Serbia.,Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Olivera Krsmanovic
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.,Armed Forces of Bosnia and Herzegovina, Logistics Command, Doboj, Bosnia and Herzegovina
| | - Nenad Solajic
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.,Department of Pathoanatomical and Laboratory Diagnostics, Oncology Institute of Vojvodina, Sremska Kamenica, Serbia
| | - Biljana Kukic
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.,Clinic for Internal Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, Serbia
| | - Ivan Nikolic
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.,Clinic for Internal Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, Serbia
| | - Bogdan Bogdanovic
- Clinic for Internal Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, Serbia
| | - Zoran Radovanovic
- Clinic for Surgical Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, Serbia.,Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Milana Kresoja
- Clinic for Surgical Oncology, Oncology Institute of Vojvodina, Sremska Kamenica, Serbia.,Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Ciaran Mannion
- Department of Pathology, Hackensack University Medical Center, Hackensack, New Jersey, USA.,Department of Pathology, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Yan-Gao Man
- Department of Pathology, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | | |
Collapse
|
13
|
Fujita K, Kamiya M, Yoshioka T, Ogasawara A, Hino R, Kojima R, Ueo H, Urano Y. Rapid and Accurate Visualization of Breast Tumors with a Fluorescent Probe Targeting α-Mannosidase 2C1. ACS CENTRAL SCIENCE 2020; 6:2217-2227. [PMID: 33376783 PMCID: PMC7760471 DOI: 10.1021/acscentsci.0c01189] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Indexed: 05/21/2023]
Abstract
Accurate detection of breast tumors and discrimination of tumor from normal tissues during breast-conserving surgery are essential to reduce the risk of misdiagnosis or recurrence. However, existing probes show substantial background signals in normal breast tissues. In this study, we focus on glycosidase activities in breast tumors. We synthesized a series of 12 fluorescent probes and performed imaging-based evaluation on surgically resected human breast specimens. Among them, the α-mannosidase-reactive fluorescent probe HMRef-αMan detected breast cancer with 90% sensitivity and 100% specificity. We identified α-mannosidase 2C1 as the target enzyme and confirmed its overexpression in various breast tumors. We found that fibroadenoma, the most common benign breast lesion in young woman, tends to have higher α-mannosidase 2C1 activity than malignant cancer. Combined application of green-emitting HMRef-αMan and a red-emitting γ-glutamyltranspeptidase probe enabled efficient dual-color, dual-target optical discrimination of malignant and benign tumors.
Collapse
Affiliation(s)
- Kyohhei Fujita
- Graduate School of Medicine and Graduate School
of Pharmaceutical Sciences, The University
of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Mako Kamiya
- Graduate School of Medicine and Graduate School
of Pharmaceutical Sciences, The University
of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
- PRESTO,
Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Takafusa Yoshioka
- Graduate School of Medicine and Graduate School
of Pharmaceutical Sciences, The University
of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Akira Ogasawara
- Graduate School of Medicine and Graduate School
of Pharmaceutical Sciences, The University
of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
| | - Rumi Hino
- Daito
Bunka University, Department of Sports and
Health Science, 560 Iwadono, Higashimatsuyama, Saitama 355-8501, Japan
| | - Ryosuke Kojima
- Graduate School of Medicine and Graduate School
of Pharmaceutical Sciences, The University
of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
- PRESTO,
Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Hiroaki Ueo
- Ueo
Breast Cancer Hospital, 1-3-5 Futamatacho, Oita, Oita 870-0887, Japan
| | - Yasuteru Urano
- Graduate School of Medicine and Graduate School
of Pharmaceutical Sciences, The University
of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo 113-0033, Japan
- CREST,
Japan
Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda,
Tokyo 100-0004, Japan
- E-mail
| |
Collapse
|
14
|
Hettie KS, Teraphongphom NT, Ertsey R, Chin FT. Off-Peak Near-Infrared-II (NIR-II) Bioimaging of an Immunoconjugate Having Peak Fluorescence Emission in the NIR-I Spectral Region for Improving Tumor Margin Delineation. ACS APPLIED BIO MATERIALS 2020; 3:8658-8666. [PMID: 35019636 PMCID: PMC9826717 DOI: 10.1021/acsabm.0c01050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The primary treatment for malignant tumors remains to be resection. The strongest predictor of recurrence and postoperative prognosis is whether diseased tissue/cells remain(s) at the surgical margin. Cancer surgery entails surgeons having the capability to visually distinguish between subtle shades of color in attempts of differentiating between diseased tissue and healthy tissue under standard white-light illumination, as such tissue states appear identical at the meso-/macroscopic level. Accordingly, enhancing the capability of surgeons to do so such that they can accurately delineate the tumor margin is of paramount importance. Fluorescence-guided surgery facilitates in enhancing such capability by color-coding the surgical field with overlaid contrasting pseudo-colors from real-time intraoperative fluorescence emission via utilizing fluorescent constructs in tandem. Constructs undergoing clinical trials or that are FDA-approved provide peak fluorescence emission in the visible (405 - 700 nm) or near-infrared-I (NIR-I) spectral region (700-900 nm), whereby differentiation between tissue states progressively improves in sync with using constructs that emit longer wavelengths of light. Here, we repurpose the usage of such fluorescent constructs by establishing feasibility of a tumor-targeting immunoconjugate (cetuximab-IRDye800) having peak fluorescence emission at the NIR-I spectral region to provide improved tumor margin delineation by affording higher tumor-to-background ratios (TBRs) when measuring its off-peak fluorescence emission at the near-infrared-II (NIR-II) spectral region (1000-1700 nm) in in vivo applications. We prepared murine tumor models, administered such immunoconjugate, and imaged such models pre-/post-administration via utilizing imaging systems that separately afforded acquisition of fluorescence emission in the NIR-I or NIR-II spectral region. On doing so, we determined in vivo TBRs, ex vivo TBRs with/-out skin, and ex vivo biodistribution, all via measuring the fluorescence emission of the immunoconjugate at tumor site(s) at both spectral regions. Collectively, we established feasibility of using the immunoconjugate to afford improved tumor margin delineation by providing 2-fold higher TBRs via utilizing the NIR-II spectral region to capture off-peak fluorescence emission from a fluorescent construct having NIR-I peak fluorescence emission.
Collapse
Affiliation(s)
- Kenneth S. Hettie
- Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, United States; Department of Otolaryngology - Head & Neck Surgery, Stanford University, Stanford, California 94305, United States
| | - Nutte Tarn Teraphongphom
- Department of Otolaryngology - Head & Neck Surgery, Stanford University, Stanford, California 94305, United States
| | - Robert Ertsey
- Department of Otolaryngology - Head & Neck Surgery, Stanford University, Stanford, California 94305, United States
| | - Frederick T. Chin
- Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
15
|
Hettie KS, Teraphongphom NT, Ertsey RD, Rosenthal EL, Chin FT. Targeting intracranial patient-derived glioblastoma (GBM) with a NIR-I fluorescent immunoconjugate for facilitating its image-guided resection. RSC Adv 2020; 10:42413-42422. [PMID: 33391732 PMCID: PMC7747479 DOI: 10.1039/d0ra07245a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/26/2020] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of primary brain tumor type and is associated with a high mortality rate borne out of such affording a survival rate of only 15 months. GBM aggressiveness is associated with the overexpression of epidermal growth factor receptor (EGFR) and its mutants. Targeting GBM with therapeutics is challenging because the blood-brain barrier (BBB) permits primarily select small-molecule entities across its semipermeable blockade. However, recent preclinical data suggest that large biomolecules, such as the anti-EGFR antibody therapeutic, cetuximab, could be capable of bypassing the BBB despite the relative enormity of its size. As such, we set forth to establish the feasibility of utilizing an EGFR-targeting near-infrared-I (NIR-I) fluorescent construct in the form of an immunoconjugate (cetuxmimab-IRDye800) to achieve visual differentiation between diseased brain tissue arising from a low-passage patient-derived GBM cell line (GBM39) and healthy brain tissue via utilizing orthotopic intracranial murine GBM39 tumor models for in vivo and ex vivo evaluation such that by doing so would establish proof of concept for ultimately facilitating its in vivo fluorescence-guided resection and ex vivo surgical back-table pathological confirmation in the clinic. As anticipated, we were not capable of distinguishing between malignant tumor tissue and healthy tissue in resected intact and slices of whole brain ex vivo under white-light illumination (WLI) due to both the diseased tissue and healthy tissue appearing virtually identical to the unaided eye. However, we readily observed over an average 6-fold enhancement in the fluorescence emission in the resected intact whole brain ex vivo when performing NIR-I fluorescence imaging (FLI) on the cohort of GBM39 tumor models that were administered the immunoconjugate compared to controls. In all, we laid the initial groundwork for establishing that NIR-I fluorescent immunoconjugates (theranostics) such as cetuximab–IRDye800 can bypass the BBB to visually afford GBM39 tumor tissue differentiation for its image-guided surgical removal. Fluorescent immunoconjugate cetuximab-IRDye800 bypasses the blood-brain-barrier to afford visualization of patient-derived GBM39 brain tumor tissue for facilitating its fluorescence-guided resection.![]()
Collapse
Affiliation(s)
- Kenneth S Hettie
- Department of Radiology, Stanford University School of Medicine, 1201 Welch Rd, Stanford, CA 94305, USA. ; Tel: +1-650-725-8172.,Department of Otolaryngology - Head & Neck Surgery, Stanford University, 1201 Welch Rd, Stanford, CA 94305, USA
| | - Nutte Tarn Teraphongphom
- Department of Otolaryngology - Head & Neck Surgery, Stanford University, 1201 Welch Rd, Stanford, CA 94305, USA
| | - Robert D Ertsey
- Department of Otolaryngology - Head & Neck Surgery, Stanford University, 1201 Welch Rd, Stanford, CA 94305, USA
| | - Eben L Rosenthal
- Department of Otolaryngology - Head & Neck Surgery, Stanford University, 1201 Welch Rd, Stanford, CA 94305, USA
| | - Frederick T Chin
- Department of Radiology, Stanford University School of Medicine, 1201 Welch Rd, Stanford, CA 94305, USA. ; Tel: +1-650-725-8172
| |
Collapse
|
16
|
Achterberg FB, Sibinga Mulder BG, Meijer RPJ, Bonsing BA, Hartgrink HH, Mieog JSD, Zlitni A, Park SM, Farina Sarasqueta A, Vahrmeijer AL, Swijnenburg RJ. Real-time surgical margin assessment using ICG-fluorescence during laparoscopic and robot-assisted resections of colorectal liver metastases. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1448. [PMID: 33313193 PMCID: PMC7723628 DOI: 10.21037/atm-20-1999] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Almost a third of the resections in patients with colorectal liver metastases (CRLM) undergoing curative surgery, end up being tumor-margin positive (≤1 mm margin). Near-infrared fluorescent (NIRF) imaging using the fluorescent contrast agent indocyanine green (ICG) has been studied for many different applications. When administered in a relatively low dose (10 mg) 24 hours prior to surgery, ICG accumulated in hepatocytes surrounding the CRLM. This results in the formation of a characteristic fluorescent 'rim' surrounding CRLM when located at the periphery of the liver. By resecting the metastasis with the entire surrounding fluorescent rim, in real-time guided by NIRF imaging, the surgeon can effectively acquire margin-negative (>1 mm) resections. This pilot study aims to describe the surgical technique for using near-infrared fluorescence imaging to assess tumor-margins in vivo in patients with CRLM undergoing laparoscopic or robot-assisted resections. Methods Out of our institutional database we selected 16 CRLM based on margin-status (R0; n=8, R1; n=8), which were resected by a minimally-invasive approach using ICG-fluorescence. NIRF images acquired during surgery, from both the resection specimen and the wound bed, were analysed for fluorescent signal. We hypothesized that a protruding fluorescent rim at the parenchymal side of the resection specimen could indicate a too close proximity to the tumor and could be predictive for a tumor-positive surgical margin. NIRF images were correlated to final histopathological assessment of the resection margin. Results All lesions with a NIRF positive resection plane in vivo were reported as having a tumor-positive margin. Lesions that showcased no protruding rim in the wound bed in vivo were diagnosed as having a tumor-negative margin in 88% of cases. A 5-step surgical workflow is described to document the NIRF signal was used assess the resection margin in vivo for future clinical studies. Conclusions The pilot study shows that image-guided surgery using real-time ICG-fluorescence has the potential to aid surgeons in achieving a tumor-negative margin in minimally invasive liver metastasectomies. The national multi-centre MIMIC-Trial will prospectively study the effect of this technique on surgical tumor-margins (Dutch Trial Register number NL7674).
Collapse
Affiliation(s)
- Friso B Achterberg
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, USA
| | | | - Ruben P J Meijer
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk H Hartgrink
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Aimen Zlitni
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, USA
| | - Seung-Min Park
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, USA
| | - Arantza Farina Sarasqueta
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Rutger-Jan Swijnenburg
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Pan J, Deng H, Hu S, Xia C, Chen Y, Wang J, Wang Y. Real-time surveillance of surgical margins via ICG-based near-infrared fluorescence imaging in patients with OSCC. World J Surg Oncol 2020; 18:96. [PMID: 32414418 PMCID: PMC7229610 DOI: 10.1186/s12957-020-01874-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/07/2020] [Indexed: 01/08/2023] Open
Abstract
Background Local recurrence is the main cause of death among patients with oral squamous cell carcinoma (OSCC). This study assessed near-infrared fluorescence (NIF) imaging and spectroscopy to monitor surgical margins intraoperatively for OSCC. Methods Cytological and animal experiments were first performed to confirm the feasibility of monitoring surgical margins with NIF imaging and spectroscopy. Then, 20 patients with OSCC were included in the clinical trials. At 6–8 h after 0.75 mg/kg indocyanine green (ICG) injection, all patients underwent surgery with NIF imaging. During the surgery, both NIF images and quantified fluorescence intensity were acquired to monitor the surgical margins. Results In cytological and animal experiments, the results showed it was feasible to monitor surgical margins with NIF imaging and spectroscopy. Fluorescence was detected in primary tumors in all patients. The fluorescence intensities of the tumor, peritumoral, and normal tissues were 398.863 ± 151.47, 278.52 ± 84.89, and 274.5 ± 100.93 arbitrary units (AUs), respectively (P < 0.05). The SBR of tumor to peritumoral tissue and normal tissues was computed to be 1.45 ± 0.36 and 1.56 ± 0.41, respectively. After primary tumor excision, the wounds showed abnormal fluorescence in four patients (4/20), and residual cancer cells were confirmed by pathological examination in two patients (2/20). Conclusion These findings confirmed the complementary value of NIF imaging during radical tumor resection of OSCC. Before tumor resection, we could utilize the fluorescence margin produced by ICG NIF imaging to determine the surgical margin. Moreover, after tumor blocks were removed, the status of surgical margin could also be evaluated rapidly by ICG NIF imaging of tumor bed and in vitro specimens.
Collapse
Affiliation(s)
- Jiongru Pan
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Han Deng
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shiqi Hu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chengwan Xia
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yongfeng Chen
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jianquan Wang
- School of Medical Imaging, Bengbu Medical College, Bengbu, China.
| | - Yuxin Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
18
|
Jensen MM, Barber ZB, Khurana N, Isaacson KJ, Steinhauff D, Green B, Cappello J, Pulsipher A, Ghandehari H, Alt JA. A dual-functional Embolization-Visualization System for Fluorescence image-guided Tumor Resection. Theranostics 2020; 10:4530-4543. [PMID: 32292513 PMCID: PMC7150499 DOI: 10.7150/thno.39700] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Intraoperative bleeding impairs physicians' ability to visualize the surgical field, leading to increased risk of surgical complications and reduced outcomes. Bleeding is particularly challenging during endoscopic-assisted surgical resection of hypervascular tumors in the head and neck. A tool that controls bleeding while marking tumor margins has the potential to improve gross tumor resection, reduce surgical morbidity, decrease blood loss, shorten procedure time, prevent damage to surrounding tissues, and limit postoperative pain. Herein, we develop and characterize a new system that combines pre-surgical embolization with improved visualization for endoscopic fluorescence image-guided tumor resection. Methods: Silk-elastinlike protein (SELP) polymers were employed as liquid embolic vehicles for delivery of a clinically used near-infrared dye, indocyanine green (ICG). The biophysical properties of SELP, including gelation kinetics, modulus of elasticity, and viscosity, in response to ICG incorporation using rheology, were characterized. ICG release from embolic SELP was modeled in tissue phantoms and via fluorescence imaging. The embolic capability of the SELP-ICG system was then tested in a microfluidic model of tumor vasculature. Lastly, the cytotoxicity of the SELP-ICG system in L-929 fibroblasts and human umbilical vein endothelial cells (HUVEC) was assessed. Results: ICG incorporation into SELP accelerated gelation and increased its modulus of elasticity. The SELP embolic system released 83 ± 8% of the total ICG within 24 hours, matching clinical practice for pre-surgical embolization procedures. Adding ICG to SELP did not reduce injectability, but did improve the gelation kinetics. After simulated embolization, ICG released from SELP in tissue phantoms diffused a sufficient distance to deliver dye throughout a tumor. ICG-loaded SELP was injectable through a clinical 2.3 Fr microcatheter and demonstrated deep penetration into 50-µm microfluidic-simulated blood vessels with durable occlusion. Incorporation of ICG into SELP improved biocompatibility with HUVECs, but had no effect on L-929 cell viability. Principle Conclusions: We report the development and characterization of a new, dual-functional embolization-visualization system for improving fluorescence-imaged endoscopic surgical resection of hypervascular tumors.
Collapse
Affiliation(s)
- M. Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Zachary B. Barber
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Nitish Khurana
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112 USA
| | - Kyle J. Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Douglas Steinhauff
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Bryant Green
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
| | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112 USA
| | - Abigail Pulsipher
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84113
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84113
| | - Jeremiah A. Alt
- Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112 USA
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84113
| |
Collapse
|
19
|
Qi B, Crawford AJ, Wojtynek NE, Talmon GA, Hollingsworth MA, Ly QP, Mohs AM. Tuned near infrared fluorescent hyaluronic acid conjugates for delivery to pancreatic cancer for intraoperative imaging. Theranostics 2020; 10:3413-3429. [PMID: 32206099 PMCID: PMC7069077 DOI: 10.7150/thno.40688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
The prognosis of pancreatic cancer remains poor. Intraoperative fluorescence imaging of tumors could improve staging and surgical resection, thereby improving prognosis. However, imaging pancreatic cancer with macromolecular delivery systems, is often hampered by nonspecific organ accumulation. Methods: We describe the rational development of hyaluronic acid (HA) conjugates that vary in molecular weight and are conjugated to near infrared fluorescent (NIRF) dyes that have differences in hydrophilicity, serum protein binding affinity, and clearance mechanism. We systematically investigated the roles of each of these properties on tumor accumulation, relative biodistribution, and the impact of intraoperative imaging of orthotopic, syngeneic pancreatic cancer. Results: Each HA-NIRF conjugate displayed intrapancreatic tumor enhancement. Regardless of HA molecular weight, Cy7.5 conjugation directed biodistribution to the liver, spleen, and bowels. Conjugation of IRDye800 to 5 and 20 kDa HA resulted in low liver and spleen signal while enhancing the tumor up to 14-fold compared to healthy pancreas, while 100 kDa HA conjugated to IRDye800 resulting in liver and spleen accumulation. Conclusion: These studies demonstrate that by tuning HA molecular weight and the physicochemical properties of the conjugated moiety, in this case a NIRF probe, peritoneal biodistribution can be substantially altered to achieve optimized delivery to tumors intraoperative abdominal imaging.
Collapse
|
20
|
Pantshwa JM, Kondiah PPD, Choonara YE, Marimuthu T, Pillay V. Nanodrug Delivery Systems for the Treatment of Ovarian Cancer. Cancers (Basel) 2020; 12:E213. [PMID: 31952210 PMCID: PMC7017423 DOI: 10.3390/cancers12010213] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Despite advances achieved in medicine, chemotherapeutics still has detrimental side effects with ovarian cancer (OC), accounting for numerous deaths among females. The provision of safe, early detection and active treatment of OC remains a challenge, in spite of improvements in new antineoplastic discovery. Nanosystems have shown remarkable progress with impact in diagnosis and chemotherapy of various cancers, due to their ideal size; improved drug encapsulation within its interior core; potential to minimize drug degradation; improve in vivo drug release kinetics; and prolong blood circulation times. However, nanodrug delivery systems have few limitations regarding its accuracy of tumour targeting and the ability to provide sustained drug release. Hence, a cogent and strategic approach has focused on nanosystem functionalization with antibody-based ligands to selectively enhance cellular uptake of antineoplastics. Antibody functionalized nanosystems are (advanced) synthetic candidates, with a broad range of efficiency in specific tumour targeting, whilst leaving normal cells unaffected. This article comprehensively reviews the present status of nanosystems, with particular emphasis on nanomicelles for molecular diagnosis and treatment of OC. In addition, biomarkers of nanosystems provide important prospects as chemotherapeutic strategies to upsurge the survival rate of patients with OC.
Collapse
Affiliation(s)
| | | | | | | | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa; (J.M.P.); (P.P.D.K.); (Y.E.C.); (T.M.)
| |
Collapse
|
21
|
Roberts S, Strome A, Choi C, Andreou C, Kossatz S, Brand C, Williams T, Bradbury M, Kircher MF, Reshetnyak YK, Grimm J, Lewis JS, Reiner T. Acid specific dark quencher QC1 pHLIP for multi-spectral optoacoustic diagnoses of breast cancer. Sci Rep 2019; 9:8550. [PMID: 31189972 PMCID: PMC6561946 DOI: 10.1038/s41598-019-44873-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/20/2019] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is the most common type of malignant growth in women. Early detection of breast cancer, as well as the identification of possible metastatic spread poses a significant challenge because of the structural and genetic heterogeneity that occurs during the progression of the disease. Currently, mammographies, biopsies and MRI scans are the standard of care techniques used for breast cancer diagnosis, all of which have their individual shortfalls, especially when it comes to discriminating tumors and benign growths. With this in mind, we have developed a non-invasive optoacoustic imaging strategy that targets the acidic environment of breast cancer. A pH low insertion peptide (pHLIP) was conjugated to the dark quencher QC1, yielding a non-fluorescent sonophore with high extinction coefficient in the near infrared that increases signal as a function of increasing amounts of membrane insertion. In an orthotopic murine breast cancer model, pHLIP-targeted optoacoustic imaging allowed us to differentiate between healthy and breast cancer tissues with high signal/noise ratios. In vivo, the sonophore QC1-pHLIP could detect malignancies at higher contrast than its fluorescent analog ICG-pHLIP, which was developed for fluorescence-guided surgical applications. PHLIP-type optoacoustic imaging agents in clinical settings are attractive due to their ability to target breast cancer and a wide variety of other malignant growths for diagnostic purposes. Intuitively, these agents could also be used for visualization during surgery.
Collapse
Affiliation(s)
- Sheryl Roberts
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Arianna Strome
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Crystal Choi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Chrysafis Andreou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Christian Brand
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Travis Williams
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Michelle Bradbury
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA.,Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, New York, 10065, USA
| | - Moritz F Kircher
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA.,Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, New York, 10065, USA.,Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.,Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA.,Department of Imaging, Dana-Farber Cancer Institute/Harvard Medical School, Boston, MA 02215, USA
| | - Yana K Reshetnyak
- Department of Physics, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Jan Grimm
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA.,Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA.,Department of Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA.,Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA.,Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA. .,Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA. .,Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, NY, 10065, United States.
| |
Collapse
|
22
|
Detection and Differentiation of Breast Cancer Sub-Types using a cPLA2α Activatable Fluorophore. Sci Rep 2019; 9:6122. [PMID: 30992473 PMCID: PMC6467920 DOI: 10.1038/s41598-019-41626-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 02/01/2019] [Indexed: 12/26/2022] Open
Abstract
Cytosolic phospholipase A2α (cPLA2α) has been shown to be elevated in breast cancer and is a potential biomarker in the differentiation of molecular sub-types. Using a cPLA2α activatable fluorophore, DDAO arachidonate, we explore its ability to function as a contrast agent in fluorescence-guided surgery. In cell lines ranging in cPLA2α expression and representing varying breast cancer sub-types, we show DDAO arachidonate activates with a high correlation to cPLA2α expression level. Using a control probe, DDAO palmitate, in addition to cPLA2α inhibition and genetic knockdown, we show that this activation is a result of cPLA2α activity. In mouse models, using an ex vivo tumor painting technique, we show that DDAO arachidonate activates to a high degree in basal-like versus luminal-like breast tumors and healthy mammary tissue. Finally, we show that using an in vivo model, orthotopic basal-like tumors give significantly high probe activation compared to healthy mammary fat pads and surrounding tissue. Together we conclude that cPLA2α activatable fluorophores such as DDAO arachidonate may serve as a useful contrast agent for the visualization of tumor margins in the fluorescence-guided surgery of basal-like breast cancer.
Collapse
|
23
|
Morris KP, Singh A, Holt DE, Stefanovski D, Singhal S, Bosco J, Capps M, McCallum M, Runge JJ. Hybrid single-port laparoscopic cisterna chyli ablation for the adjunct treatment of chylothorax disease in dogs. Vet Surg 2019; 48:O121-O129. [PMID: 30927322 DOI: 10.1111/vsu.13195] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To describe a hybrid, single-port, minimally invasive cisterna chyli ablation (CCA) technique in clinical cases of canine idiopathic chylothorax and evaluate this technique as a method for mesenteric lymphangiography (ML) in canine cadavers and clinical cases of idiopathic chylothorax. STUDY DESIGN Cadaveric and retrospective study. ANIMALS Six canine cadavers and 14 client-owned dogs with naturally occurring idiopathic chylothorax. METHODS Both cadaveric and clinically affected dogs were placed in sternal recumbency. A wound retractor device (WRD) and a single-port device were placed in the abdominal flank 2-3 cm caudal to the 13th rib. Mesenteric lymphangiography was evaluated by using indocyanine green (ICG) in 6 canine cadavers. Single-port laparoscopic CCA was performed in all clinical cases with idiopathic chylothorax. RESULTS Successful ML was completed by using ICG in all 6 canine cadavers. A right- or left-sided single-port laparoscopic CCA was successfully performed in 14 dogs with naturally occurring idiopathic chylothorax. Mesenteric lymphangiography was successfully performed through the WRD in 11 of these cases. No intraoperative complications were reported. Three dogs developed severe chyloabdomen postoperatively, with 1 dog requiring multiple abdominocenteses. CONCLUSION Direct ML and single-port laparoscopic CCA was performed through a WRD in dogs positioned in sternal recumbency. Although minimal operative complications were noted, postoperative chyloabdomen was reported. CLINICAL SIGNIFICANCE This hybrid single-port laparoscopic technique performed in sternal recumbency allows both a CCA and an intraoperative ML through the same incision. This procedure may be combined with thoracoscopic thoracic duct ligation and pericardectomy for the treatment of idiopathic chylothorax in dogs.
Collapse
Affiliation(s)
- Katherine P Morris
- Department of Clinical Studies, Section of Surgery, Veterinary Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ameet Singh
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - David E Holt
- Department of Clinical Studies, Section of Surgery, Veterinary Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Darko Stefanovski
- Department of Biostatistics, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Jessica Bosco
- Department of Clinical Studies, Section of Surgery, Veterinary Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michelle Capps
- Department of Clinical Studies, Section of Surgery, Veterinary Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael McCallum
- Department of Clinical Studies, Section of Surgery, Veterinary Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey J Runge
- Department of Surgery, Guardian Veterinary Specialists, Brewster, New York
| |
Collapse
|
24
|
Chung HY, Greinert R, Kärtner FX, Chang G. Multimodal imaging platform for optical virtual skin biopsy enabled by a fiber-based two-color ultrafast laser source. BIOMEDICAL OPTICS EXPRESS 2019; 10:514-525. [PMID: 30800496 PMCID: PMC6377886 DOI: 10.1364/boe.10.000514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 05/07/2023]
Abstract
We demonstrate multimodal label-free nonlinear optical microscopy in human skin enabled by a fiber-based two-color ultrafast source. Energetic femtosecond pulses at 775 nm and 1250 nm are simultaneously generated by an Er-fiber laser source employing frequency doubling and self-phase modulation enabled spectral selection. The integrated nonlinear optical microscope driven by such a two-color femtosecond source enables the excitation of endogenous two-photon excitation fluorescence, second-harmonic generation, and third-harmonic generation in human skin. Such a 3-channel imaging platform constitutes a powerful tool for clinical application and optical virtual skin biopsy.
Collapse
Affiliation(s)
- Hsiang-Yu Chung
- Center for Free-Electron Laser Science, DESY, Notkestraße 85, 22607 Hamburg, Germany
- Physics Department, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
| | | | - Franz X Kärtner
- Center for Free-Electron Laser Science, DESY, Notkestraße 85, 22607 Hamburg, Germany
- Physics Department, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
- The Hamburg Centre for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
| | - Guoqing Chang
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
25
|
Wang Y, Xie D, Wang Z, Zhang X, Zhang Q, Wang Y, Newton AD, Singhal S, Cai H, Wang Y, Lu Q, Hu Q, Wang Z. Kinetics of indocyanine green: Optimizing tumor to normal tissue fluorescence in image-guided oral cancer surgery applications. Head Neck 2018; 41:1032-1038. [PMID: 30549410 DOI: 10.1002/hed.25541] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 08/08/2018] [Accepted: 10/05/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The study aimed to define indocyanine green (ICG) kinetics to determine the optimal ICG dose and surgical time for near-infrared fluorescence-guided oral cancer surgery. METHODS Spectrometer and grayscale digital imaging were used to quantify the ICG kinetics in 12 patients with oral cancer. The fluorescence intensity and signal-to-background ratio (SBR) of tumor and normal tissue were tested at 1, 6, and 24 hours after ICG injection. RESULTS The greatest contrast in the fluorescence intensity between tumor and normal tissue was observed at 6 hours (P < .01), and of three dose groups (0.5, 0.75, and 1.0 mg/kg), 0.75 mg/kg showed the highest SBR (2.06 ± 0.23) after ICG injection. CONCLUSIONS Fluorescence quantification based on spectrometry and grayscale imaging could be effective in determining the optimal ICG dose and surgical time after ICG injection in this cohort of patients with oral cancer.
Collapse
Affiliation(s)
- Yuxin Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Diya Xie
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ziyang Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Xudong Zhang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Qian Zhang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yang Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Andrew D Newton
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Huiming Cai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Yiqing Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Qian Lu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhiyong Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
26
|
Predina JD, Okusanya O, D Newton A, Low P, Singhal S. Standardization and Optimization of Intraoperative Molecular Imaging for Identifying Primary Pulmonary Adenocarcinomas. Mol Imaging Biol 2018; 20:131-138. [PMID: 28497233 DOI: 10.1007/s11307-017-1076-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Intraoperative molecular imaging (IMI) is an emerging technology used to locate pulmonary adenocarcinomas and identify positive margins during surgery. Background noise and tissue autofluorescence have been major obstacles. The goal of this study is to optimize the image quality of folate receptor alpha (FRα) targeted IMI for pulmonary adenocarcinomas by modifying emission data. PROCEDURES A total of 15 lung cancer patients were enrolled in a pilot study. In the first cohort, FRα upregulation within pulmonary adenocarcinoma tumors was confirmed by analyzing specimens from five pulmonary adenocarcinoma patients with flow cytometry and immunohistochemistry. Next, in a cohort of five additional patients, autofluorescence of intrathoracic structures and tissues was quantified. Lastly, five patients with tumors at various depths from the pleural surface were enrolled and received the FRα-targeted optical contrast agent, EC17. In this final cohort, resected pulmonary adenocarcinomas were imaged at a wide range of fluorescence exposure times (0 to 200 ms), various laser powers, and with unique filter configurations. Tumor-to-noise ratio (TNR) for images was generated using region of interest software. RESULTS Pulmonary adenocarcinomas highly express FRα. Significant autofluorescence from native thoracic tissues was found with the highest fluorescent signals at the bronchial stump (547 ± 98, range 423-699), the pulmonary artery (267 ± 64, range 200-374), and cortical bone (266 ± 17, range 243-287). High levels of autofluorescence were appreciated after systemic administration of EC17; however, TNR was improved by altering exposure settings at the time of the imaging. Optimal fluorescent exposure time occurs at 40 ms (25 frames/s). CONCLUSIONS Exposure properties can be manipulated to maximize TNR thus allowing for successful intraoperative detection of pulmonary adenocarcinomas during surgery. Optimization of the conditions for intraoperative molecular imaging sets the stage for future clinical trials utilizing targeted IMI techniques which can aid the surgeon at the time of cancer resection.
Collapse
Affiliation(s)
- Jarrod D Predina
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Olugbenga Okusanya
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Andrew D Newton
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA.,Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Philip Low
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Sunil Singhal
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA. .,Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
27
|
Banerjee S. Ambient ionization mass spectrometry imaging for disease diagnosis: Excitements and challenges. J Biosci 2018; 43:731-738. [PMID: 30207318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Tissue analysis in histology is extremely important and also considered to be a gold standard to diagnose and prognosticate several diseases including cancer. Intraoperative evaluation of surgical margin of tumor also relies on frozen section histopathology, which is time consuming, challenging and often subjective. Recent development in the ambient ionization mass spectrometry imaging (MSI) technique has enabled us to simultaneously visualize hundreds to thousands of molecules (ion images) in the biopsy specimen, which are strikingly different and more powerful than the single optical tissue image analysis in conventional histopathology. This paper will highlight the emergence of the desorption electrospray ionization MSI (DESI-MSI) technique, which is label-free, requires minimal or no sample preparation and operates under ambient conditions. DESI-MSI can record ion images of lipid/metabolite distributions on biopsy specimens, providing a wealth of diagnostic information based on differential distributions of these molecular species in healthy and unhealthy tissues. Remarkable success of this technology in rapidly evaluating the cancer margin intraoperatively with very high accuracy also promises to bring this imaging technique from bench to bedside.
Collapse
Affiliation(s)
- Shibdas Banerjee
- Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Karakambadi Road, Tirupati 517 507, India,
| |
Collapse
|
28
|
Banerjee S. Ambient ionization mass spectrometry imaging for disease diagnosis: Excitements and challenges. J Biosci 2018. [DOI: 10.1007/s12038-018-9785-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Low PS, Singhal S, Srinivasarao M. Fluorescence-guided surgery of cancer: applications, tools and perspectives. Curr Opin Chem Biol 2018; 45:64-72. [PMID: 29579618 DOI: 10.1016/j.cbpa.2018.03.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 12/20/2022]
Abstract
Thousands of patients die each year from residual cancer that remains following cytoreductive surgery. Use of tumor-targeted fluorescent dyes (TTFDs) to illuminate undetected malignant tissue and thereby facilitate its surgical resection shows promise for reducing morbidity and mortality associated with unresected malignant disease. TTFDs can also improve i) detection of recurrent malignant lesions, ii) differentiation of normal from malignant lymph nodes, iii) accurate staging of cancer patients, iv) detection of tumors during robotic/endoscopic surgery (where tumor palpation is no longer possible), and v) preservation of healthy tissue during resection of cancer tissue. Although TTFDs that passively accumulate in a tumor mass provide some tumor contrast, the most encouraging TTFDs in human clinical trials are either enzyme-activated or ligand-targeted to tumor-specific receptors.
Collapse
Affiliation(s)
- Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States.
| | - Sunil Singhal
- Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
30
|
Bernthal NM. CORR Insights®: Quantitative Primary Tumor Indocyanine Green Measurements Predict Osteosarcoma Metastatic Lung Burden in a Mouse Model. Clin Orthop Relat Res 2018; 476. [PMID: 29529629 PMCID: PMC6260027 DOI: 10.1007/s11999.0000000000000136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
31
|
Fourman MS, Mahjoub A, Mandell JB, Yu S, Tebbets JC, Crasto JA, Alexander PE, Weiss KR. Quantitative Primary Tumor Indocyanine Green Measurements Predict Osteosarcoma Metastatic Lung Burden in a Mouse Model. Clin Orthop Relat Res 2018; 476:479-487. [PMID: 29408832 PMCID: PMC6260021 DOI: 10.1007/s11999.0000000000000003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Current preclinical osteosarcoma (OS) models largely focus on quantifying primary tumor burden. However, most fatalities from OS are caused by metastatic disease. The quantification of metastatic OS currently relies on CT, which is limited by motion artifact, requires intravenous contrast, and can be technically demanding in the preclinical setting. We describe the ability for indocyanine green (ICG) fluorescence angiography to quantify primary and metastatic OS in a previously validated orthotopic, immunocompetent mouse model. QUESTIONS/PURPOSES (1) Can near-infrared ICG fluorescence be used to attach a comparable, quantitative value to the primary OS tumor in our experimental mouse model? (2) Will primary tumor fluorescence differ in mice that go on to develop metastatic lung disease? (3) Does primary tumor fluorescence correlate with tumor volume measured with CT? METHODS Six groups of 4- to 6-week-old immunocompetent Balb/c mice (n = 6 per group) received paraphyseal injections into their left hindlimb proximal tibia consisting of variable numbers of K7M2 mouse OS cells. A hindlimb transfemoral amputation was performed 4 weeks after injection with euthanasia and lung extraction performed 10 weeks after injection. Histologic examination of lung and primary tumor specimens confirmed ICG localization only within the tumor bed. RESULTS Mice with visible or palpable tumor growth had greater hindlimb fluorescence (3.5 ± 2.3 arbitrary perfusion units [APU], defined as the fluorescence pixel return normalized by the detector) compared with those with a negative examination (0.71 ± 0.38 APU, -2.7 ± 0.5 mean difference, 95% confidence interval -3.7 to -1.8, p < 0.001). A strong linear trend (r = 0.81, p < 0.01) was observed between primary tumor and lung fluorescence, suggesting that quantitative ICG tumor fluorescence is directly related to eventual metastatic burden. We did not find a correlation (r = 0.04, p = 0.45) between normalized primary tumor fluorescence and CT volumetric measurements. CONCLUSIONS We demonstrate a novel methodology for quantifying primary and metastatic OS in a previously validated, immunocompetent, orthotopic mouse model. Quantitative fluorescence of the primary tumor with ICG angiography is linearly related to metastatic burden, a relationship that does not exist with respect to clinical tumor size. This highlights the potential utility of ICG near-infrared fluorescence imaging as a valuable preclinical proof-of-concept modality. Future experimental work will use this model to evaluate the efficacy of novel OS small molecule inhibitors. CLINICAL RELEVANCE Given the histologic localization of ICG to only the tumor bed, we envision the clinical use of ICG angiography as an intraoperative margin and tumor detector. Such a tool may be used as an alternative to intraoperative histology to confirm negative primary tumor margins or as a valuable tool for debulking surgeries in vulnerable anatomic locations. Because we have demonstrated the successful preservation of ICG in frozen tumor samples, future work will focus on blinded validation of this modality in observational human trials, comparing the ICG fluorescence of harvested tissue samples with fresh frozen pathology.
Collapse
Affiliation(s)
- Mitchell S Fourman
- M. S. Fourman, J. B. Mandell, S. Yu, J. C. Tebbets, J. A. Crasto, K. R. Weiss Cancer Stem Cell Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA A. Mahjoub School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; and the Cancer Stem Cell Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA P. E. Alexander Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Wu MR, Liu HM, Lu CW, Shen WH, Lin IJ, Liao LW, Huang YY, Shieh MJ, Hsiao JK. Organic anion-transporting polypeptide 1B3 as a dual reporter gene for fluorescence and magnetic resonance imaging. FASEB J 2018; 32:1705-1715. [PMID: 29146731 PMCID: PMC5892727 DOI: 10.1096/fj.201700767r] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Reporter proteins have broad applications in visualizing molecular events at the cellular, tissue and whole-body levels. Transmembrane transporters recognizing specific molecular domains are of particular interest because they enable the migration of signal-source molecules from the extracellular space to the cytoplasm for subsequent application in multimodality imaging. Organic anion-transporting polypeptides (OATPs) have demonstrated their MRI reporter efficacy. We further expanded their use as a dual-modality reporter in MRI and noninvasive in vivo imaging system (IVIS). We overexpressed OATP1B3 in the HT-1080 sarcoma cell line. Both Gd-EOB-DTPA, an MRI contrast agent, and indocyanine green (ICG), a near-infrared fluorescent dye that provides better deep-tissue detection because of its long wavelength, could be delivered to the intracellular space and imaged in a tumor-bearing nude mouse model. Our in vivo dual-imaging reporter system achieved high sensitivity in MRI and observation periods lasting as long as 96 h in IVIS. Because of the superior temporal and spatial resolutions and the clinical availability of both ICG and Gd-EOB-DTPA, this dual-imaging OATP1B3 system will find biomedical use in tumor biology, stem cell trafficking, and tissue engineering.—Wu, M.-R., Liu, H.-M., Lu, C.-W., Shen, W.-H., Lin, I.-J., Liao, L.-W., Huang, Y.-Y., Shieh, M.-J., Hsiao, J.-K. Organic anion-transporting polypeptide 1B3 as a dual reporter gene for fluorescence and magnetic resonance imaging.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Hon-Man Liu
- Department of Medical Imaging, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan.,Department of Radiology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Radiology and Medical Imaging, Fu-Jen Catholic University and Hospital, New Taipei City, Taiwan
| | - Chen-Wen Lu
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan.,Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Way-Hone Shen
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - I-Jou Lin
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Li-Wen Liao
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
33
|
He K, Zhou J, Yang F, Chi C, Li H, Mao Y, Hui B, Wang K, Tian J, Wang J. Near-infrared Intraoperative Imaging of Thoracic Sympathetic Nerves: From Preclinical Study to Clinical Trial. Theranostics 2018; 8:304-313. [PMID: 29290809 PMCID: PMC5743549 DOI: 10.7150/thno.22369] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/02/2017] [Indexed: 12/15/2022] Open
Abstract
The sympathetic nervous system controls and regulates the activities of the heart and other organs. Sympathetic nervous system dysfunction leads to disease. Therefore, intraoperative real-time imaging of thoracic sympathetic nerves (ITSN) would be of great clinical significance for diagnosis and therapy. The aim of this experimental study was to evaluate the feasibility and validity of intraoperative ITSN using indocyanine green (ICG). METHODS ITSN using ICG was performed on 10 rabbits to determine its feasibility. Animals were allocated to two groups. The rabbits in one group received the same dose of ICG, but were observed at different times. The rabbits in the other group were administered different doses of ICG, but were observed at the same time. Signal to background ratio (SBR) was measured in regions of interest in all rabbits. Furthermore, fifteen consecutive patients with pulmonary nodules were intravenously injected with ICG 24 h preoperatively and underwent near-infrared (NIR) fluorescence imaging (FI) thoracoscopic surgeries between July 2015 and June 2016. A novel self-developed NIR and white-light dual-channel thoracoscope system was used. SBRs of thoracic sympathetic nerves were calculated in all patients. RESULTS In the preclinical study, we were able to precisely recognize each rabbit's second (T2) to fifth (T5) thoracic ganglia on both sides of the spine using ITSN with ICG. In addition, we explored the relationship between SBR and the injection time of ICG and that between SBR and the dose of ICG. Using the novel dual-channel thoracoscope system, we were able to locate the ganglia from the stellate ganglion (SG) to the sixth thoracic ganglion (T6), as well as the chains between these ganglia in all patients with a high SBR value of 3.26 (standard deviation: 0.57). The pathological results confirmed our findings. CONCLUSION We were able to use ICG FI to distinguish thoracic sympathetic nerves during NIR thoracoscopic surgery. The technique may replace the rib-oriented method as standard practice for mapping the thoracic sympathetic nerves.
Collapse
Affiliation(s)
- Kunshan He
- Department of Thoracic Surgery, Peking University People's Hospital, No.11, Xi Zhi Men South Avenue, Beijing100190, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Jian Zhou
- Department of Thoracic Surgery, Peking University People's Hospital, No.11, Xi Zhi Men South Avenue, Beijing100190, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, No.11, Xi Zhi Men South Avenue, Beijing100190, China
| | - Chongwei Chi
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Hao Li
- Department of Thoracic Surgery, Peking University People's Hospital, No.11, Xi Zhi Men South Avenue, Beijing100190, China
| | - Yamin Mao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Bengang Hui
- Department of Thoracic Surgery, Peking University People's Hospital, No.11, Xi Zhi Men South Avenue, Beijing100190, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100039, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jun Wang
- Department of Thoracic Surgery, Peking University People's Hospital, No.11, Xi Zhi Men South Avenue, Beijing100190, China
| |
Collapse
|
34
|
Sönnichsen R, Hennig L, Blaschke V, Winter K, Körfer J, Hähnel S, Monecke A, Wittekind C, Jansen-Winkeln B, Thieme R, Gockel I, Grosser K, Weimann A, Kubick C, Wiechmann V, Aigner A, Bechmann I, Lordick F, Kallendrusch S. Individual Susceptibility Analysis Using Patient-derived Slice Cultures of Colorectal Carcinoma. Clin Colorectal Cancer 2017; 17:e189-e199. [PMID: 29233603 DOI: 10.1016/j.clcc.2017.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/18/2017] [Accepted: 11/14/2017] [Indexed: 02/01/2023]
Abstract
BACKGROUND Nonresponse to chemotherapy in colorectal carcinoma (CRC) is still a clinical problem. For most established treatment regimens, no predictive biomarkers are available. Patient-derived tumor slice culture may be a promising ex vivo technology to assess the drug susceptibility in individual tumors. METHODS Patient-derived slice cultures of CRC specimens were prepared according to a standardized protocol and treated with different concentrations of 5-fluorouracil (5-FU) and an adapted FOLFOX regimen (5-FU and oxaliplatin) to investigate histologic response. Additionally, a semi-automatized readout using fluorescent stain-specific segmentation algorithms for Image J was established to quantify changes in tumor proliferation. Nonresponse to chemotherapy was defined as persisting tumor cell proliferation. RESULTS Slices treated with 5-FU showed lower tumor cell fractions and dose-dependent alterations of proliferating tumor cells compared with controls (1 μM, Δ +3%; 10 μM, Δ -9%; 100 μM, Δ -15%). Individual tumor samples were examined and differences in chemotherapy susceptibility could be observed. Untreated slice cultures contained an average tumor cell fraction of 31% ± 7%. For all samples, the histopathologic characteristics exhibited some degree of intratumoral heterogeneity with regard to tumor cell morphology and distribution. The original tumor matched the features found in slices at baseline and after 3 days of cultivation. CONCLUSIONS Patient-derived slice cultures may help to predict response to clinical treatment in individual patients with CRC. Future studies need to address the problem of tumor heterogeneity and evolution. Prospective correlation of ex vivo results with the clinical course of treated patients is warranted.
Collapse
Affiliation(s)
- Rasmus Sönnichsen
- Institute of Anatomy, University of Leipzig, Leipzig, Germany; University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany.
| | - Laura Hennig
- Institute of Anatomy, University of Leipzig, Leipzig, Germany; University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | - Vera Blaschke
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Justus Körfer
- Institute of Anatomy, University of Leipzig, Leipzig, Germany; University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | - Susann Hähnel
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Astrid Monecke
- Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
| | | | - Boris Jansen-Winkeln
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - René Thieme
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Ines Gockel
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Grosser
- Department of General and Visceral Surgery, St. Georg Hospital, Leipzig, Germany
| | - Arved Weimann
- Department of General and Visceral Surgery, St. Georg Hospital, Leipzig, Germany
| | | | | | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Florian Lordick
- University Cancer Center Leipzig, University Hospital Leipzig, Leipzig, Germany
| | | |
Collapse
|
35
|
Arlauckas SP, Kumar M, Popov AV, Poptani H, Delikatny EJ. Near infrared fluorescent imaging of choline kinase alpha expression and inhibition in breast tumors. Oncotarget 2017; 8:16518-16530. [PMID: 28157707 PMCID: PMC5369982 DOI: 10.18632/oncotarget.14965] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
Choline kinase alpha (ChoKα) overexpression is associated with an aggressive tumor phenotype. ChoKα inhibitors induce apoptosis in tumors, however validation of their specificity is difficult in vivo. We report the use of optical imaging to assess ChoKα status in cells and in vivo using JAS239, a carbocyanine-based ChoKα inhibitor with inherent near infrared fluorescence. JAS239 attenuated choline phosphorylation and viability in a panel of human breast cancer cell lines. Antibody blockade prevented cellular retention of JAS239 indicating direct interaction with ChoKα independent of the choline transporters and catabolic choline pathways. In mice bearing orthotopic MCF7 breast xenografts, optical imaging with JAS239 distinguished tumors overexpressing ChoKα from their empty vector counterparts and delineated tumor margins. Pharmacological inhibition of ChoK by the established inhibitor MN58b led to a growth inhibition in 4175-Luc+ tumors that was accompanied by concomitant reduction in JAS239 uptake and decreased total choline metabolite levels as measured using magnetic resonance spectroscopy. At higher therapeutic doses, JAS239 was as effective as MN58b at arresting tumor growth and inducing apoptosis in MDA-MB-231 tumors, significantly reducing tumor choline below baseline levels without observable systemic toxicity. These data introduce a new method to monitor therapeutically effective inhibitors of choline metabolism in breast cancer using a small molecule companion diagnostic.
Collapse
Affiliation(s)
- Sean P Arlauckas
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Manoj Kumar
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anatoliy V Popov
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Harish Poptani
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Department of Cellular and Molecular Physiology, Institute of Regenerative Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Edward J Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
36
|
Banerjee S, Zare RN, Tibshirani RJ, Kunder CA, Nolley R, Fan R, Brooks JD, Sonn GA. Diagnosis of prostate cancer by desorption electrospray ionization mass spectrometric imaging of small metabolites and lipids. Proc Natl Acad Sci U S A 2017; 114:3334-3339. [PMID: 28292895 PMCID: PMC5380053 DOI: 10.1073/pnas.1700677114] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Accurate identification of prostate cancer in frozen sections at the time of surgery can be challenging, limiting the surgeon's ability to best determine resection margins during prostatectomy. We performed desorption electrospray ionization mass spectrometry imaging (DESI-MSI) on 54 banked human cancerous and normal prostate tissue specimens to investigate the spatial distribution of a wide variety of small metabolites, carbohydrates, and lipids. In contrast to several previous studies, our method included Krebs cycle intermediates (m/z <200), which we found to be highly informative in distinguishing cancer from benign tissue. Malignant prostate cells showed marked metabolic derangements compared with their benign counterparts. Using the "Least absolute shrinkage and selection operator" (Lasso), we analyzed all metabolites from the DESI-MS data and identified parsimonious sets of metabolic profiles for distinguishing between cancer and normal tissue. In an independent set of samples, we could use these models to classify prostate cancer from benign specimens with nearly 90% accuracy per patient. Based on previous work in prostate cancer showing that glucose levels are high while citrate is low, we found that measurement of the glucose/citrate ion signal ratio accurately predicted cancer when this ratio exceeds 1.0 and normal prostate when the ratio is less than 0.5. After brief tissue preparation, the glucose/citrate ratio can be recorded on a tissue sample in 1 min or less, which is in sharp contrast to the 20 min or more required by histopathological examination of frozen tissue specimens.
Collapse
Affiliation(s)
- Shibdas Banerjee
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Richard N Zare
- Department of Chemistry, Stanford University, Stanford, CA 94305;
| | - Robert J Tibshirani
- Department of Biomedical Data Sciences, Stanford University, Stanford, CA 94305
- Department of Statistics, Stanford University, Stanford, CA 94305
| | - Christian A Kunder
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Rosalie Nolley
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305
| | - Richard Fan
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305
| | - Geoffrey A Sonn
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
37
|
Mondal SB, Gao S, Zhu N, Habimana-Griffin L, Akers WJ, Liang R, Gruev V, Margenthaler J, Achilefu S. Optical See-Through Cancer Vision Goggles Enable Direct Patient Visualization and Real-Time Fluorescence-Guided Oncologic Surgery. Ann Surg Oncol 2017; 24:1897-1903. [PMID: 28213790 DOI: 10.1245/s10434-017-5804-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND The inability to visualize the patient and surgical site directly, limits the use of current near infrared fluorescence-guided surgery systems for real-time sentinel lymph node biopsy and tumor margin assessment. METHODS We evaluated an optical see-through goggle augmented imaging and navigation system (GAINS) for near-infrared, fluorescence-guided surgery. Tumor-bearing mice injected with a near infrared cancer-targeting agent underwent fluorescence-guided, tumor resection. Female Yorkshire pigs received hind leg intradermal indocyanine green injection and underwent fluorescence-guided, popliteal lymph node resection. Four breast cancer patients received 99mTc-sulfur colloid and indocyanine green retroareolarly before undergoing sentinel lymph node biopsy using radioactive tracking and fluorescence imaging. Three other breast cancer patients received indocyanine green retroareolarly before undergoing standard-of-care partial mastectomy, followed by fluorescence imaging of resected tumor and tumor cavity for margin assessment. RESULTS Using near-infrared fluorescence from the dyes, the optical see-through GAINS accurately identified all mouse tumors, pig lymphatics, and four pig popliteal lymph nodes with high signal-to-background ratio. In 4 human breast cancer patients, 11 sentinel lymph nodes were identified with a detection sensitivity of 86.67 ± 0.27% for radioactive tracking and 100% for GAINS. Tumor margin status was accurately predicted by GAINS in all three patients, including clear margins in patients 1 and 2 and positive margins in patient 3 as confirmed by paraffin-embedded section histopathology. CONCLUSIONS The optical see-through GAINS prototype enhances near infrared fluorescence-guided surgery for sentinel lymph node biopsy and tumor margin assessment in breast cancer patients without disrupting the surgical workflow in the operating room.
Collapse
Affiliation(s)
- Suman B Mondal
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Shengkui Gao
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Nan Zhu
- College of Optical Science, The University of Arizona, Tucson, AZ, USA
| | - LeMoyne Habimana-Griffin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Walter J Akers
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rongguang Liang
- College of Optical Science, The University of Arizona, Tucson, AZ, USA
| | - Viktor Gruev
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie Margenthaler
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA. .,Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA. .,Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
38
|
Keating J, Newton A, Venegas O, Nims S, Zeh R, Predina J, Deshpande C, Kucharczuk J, Nie S, Delikatny EJ, Singhal S. Near-Infrared Intraoperative Molecular Imaging Can Locate Metastases to the Lung. Ann Thorac Surg 2017; 103:390-398. [PMID: 27793401 PMCID: PMC11024498 DOI: 10.1016/j.athoracsur.2016.08.079] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pulmonary metastasectomy is widely accepted for many tumor types because it may prolong survival and potentially cure some patients. However, intraoperative localization of pulmonary metastases can be technically challenging. We propose that intraoperative near-infrared (NIR) molecular imaging can be used as an adjunct during disease localization. METHODS We inoculated 50 C57BL/6 mice with Lewis lung carcinoma (LLC) flank tumors. After flank tumor growth, mice were injected through the tail vein with indocyanine green (ICG) before operation, and intraoperative imaging was used to detect pulmonary metastases. On the basis of these experiments, we enrolled 8 patients undergoing pulmonary metastasectomy into a pilot and feasibility clinical trial. Each patient received intravenous ICG 1 day before operation, followed by wedge or segmental resection. Samples were imaged on the back table with an NIR camera to confirm disease presence and margins. All murine and human tumors and margins were confirmed by pathologic examination. RESULTS Mice had an average of 4 ± 2 metastatic tumors on both lungs, with an average size of 5.1 mm (interquartile range [IQR] 2.2 mm to 7.6 mm). Overall, 200 of 211 (95%) metastatic deposits were markedly fluorescent, with a mean tumor-to-background ratio (TBR) of 3.4 (IQR 3.1 to 4.1). The remaining tumors had a TBR below 1.5. In the human study, intraoperative NIR imaging identified six of the eight preoperatively localized lesions. Intraoperative back table NIR imaging identified all metastatic lesions, which were confirmed by pathologic examination. The average tumor size was 1.75 ± 1.4 cm, and the mean ex vivo TBR was 3.3 (IQR 3.1 to 3.7). Pathologic examination demonstrated melanoma (n = 4), osteogenic sarcoma (n = 2), renal cell carcinoma (n = 2), chondrosarcoma (n = 1), leiomyosarcoma (n = 1), and colorectal carcinoma (n = 1). CONCLUSIONS Systemic ICG identifies subcentimeter tumor metastases to the lung in murine models, and this work provides proof of principle in humans. Future research is focused on improving depth of penetration into the lung parenchyma.
Collapse
Affiliation(s)
- Jane Keating
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Andrew Newton
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ollin Venegas
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sarah Nims
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ryan Zeh
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jarrod Predina
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Charuhas Deshpande
- Department of Pathology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - John Kucharczuk
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shuming Nie
- Departments of Biomedical Engineering and Chemistry, Emory University, Atlanta, Georgia
| | - E James Delikatny
- Department of Radiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
39
|
Griffin JI, Benchimol MJ, Simberg D. Longitudinal monitoring of skin accumulation of nanocarriers and biologicals with fiber optic near infrared fluorescence spectroscopy (FONIRS). J Control Release 2017; 247:167-174. [PMID: 28069552 DOI: 10.1016/j.jconrel.2017.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/21/2016] [Accepted: 01/04/2017] [Indexed: 01/04/2023]
Abstract
Systemically injected drug delivery systems distribute into various organs and tissues, including liver, spleen and kidneys. Recent reports pointed out a significant accumulation of systemically injected nanoparticles in the skin. Skin constitutes the largest organ in the body with important immune functions, and accumulation of drug delivery systems could have significant implications for skin toxicity in living subjects. Fiber optic-based near-infrared spectroscopy (FONIRS) setup was developed and tested for measuring of NIR (760nm excitation) emission spectra in the skin. Ex vivo spectral measurements of NIR fluorescence through the skin showed linear response down to 34 femtomole of dye DiR. Following systemic injection of IRDye 800 labeled 500kDa dextran, FONIRS detected an immediate and stable accumulation of fluorescence in the skin. Longitudinal monitoring of skin accumulation and elimination of IRDye 800-labeled therapeutic anti-epidermal growth factor antibody (cetuximab) showed significant signal in the skin after the antibody cleared from circulation. Comparison of skin accumulation of DiR labeled, long-circulating PEGylated liposomes with short-circulating non-PEGylated liposomes showed much higher accumulation of PEGylated liposomes that persisted several days after the liposomes cleared from blood. Measurements with FONIRS enabled to estimate skin concentration of liposomes (percent of injected dose per gram). This simple and practical approach can be used to monitor accumulation of drug delivery systems in preclinical and clinical studies.
Collapse
Affiliation(s)
- James I Griffin
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd., Aurora, CO 80045, USA
| | | | - Dmitri Simberg
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 East Montview Blvd., Aurora, CO 80045, USA.
| |
Collapse
|
40
|
Kairdolf BA, Qian X, Nie S. Bioconjugated Nanoparticles for Biosensing, in Vivo Imaging, and Medical Diagnostics. Anal Chem 2017; 89:1015-1031. [DOI: 10.1021/acs.analchem.6b04873] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Brad A. Kairdolf
- Department of Biomedical
Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
| | - Ximei Qian
- Department of Biomedical
Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
| | - Shuming Nie
- Department of Biomedical
Engineering, Emory University and Georgia Institute of Technology, 1760 Haygood Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
41
|
Abstract
Indocyanine green (ICG) is the only near-infrared dye approved by the U.S. Food and Drug Administration for clinical use. When injected in blood, ICG binds primarily to plasma proteins and lipoproteins, resulting in enhanced fluorescence. Recently, the optofluidic laser has emerged as a novel tool in bio-analysis. Laser emission has advantages over fluorescence in signal amplification, narrow linewidth, and strong intensity, leading to orders of magnitude increase in detection sensitivity and imaging contrast. Here we successfully demonstrate, to the best of our knowledge, the first ICG lasing in human serum and whole blood with the clinical ICG concentrations and the pump intensity far below the clinically permissible level. Furthermore, we systematically study ICG laser emission within each major serological component (albumins, globulins, and lipoproteins) and reveal the critical elements and conditions responsible for lasing. Our work marks a critical step toward eventual clinical and biomedical applications of optofluidic lasers using FDA approved fluorophores, which may complement or even supersede conventional fluorescence-based sensing and imaging.
Collapse
Affiliation(s)
- Yu-Cheng Chen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, 1101 Beal Ave., Ann Arbor, Michigan 48109, USA
| | - Qiushu Chen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, 1101 Beal Ave., Ann Arbor, Michigan 48109, USA
| | - Xudong Fan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, 1101 Beal Ave., Ann Arbor, Michigan 48109, USA
| |
Collapse
|
42
|
Ankri R, Ashkenazy A, Milstein Y, Brami Y, Olshinka A, Goldenberg-Cohen N, Popovtzer A, Fixler D, Hirshberg A. Gold Nanorods Based Air Scanning Electron Microscopy and Diffusion Reflection Imaging for Mapping Tumor Margins in Squamous Cell Carcinoma. ACS NANO 2016; 10:2349-56. [PMID: 26759920 DOI: 10.1021/acsnano.5b07114] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
A critical challenge arising during a surgical procedure for tumor removal is the determination of tumor margins. Gold nanorods (GNRs) conjugated to epidermal growth factor receptors (EGFR) (GNRs-EGFR) have long been used in the detection of cancerous cells as the expression of EGFR dramatically increases once the tissue becomes cancerous. Optical techniques for the identification of these GNRs-EGFR in tumor are intensively developed based on the unique scattering and absorption properties of the GNRs. In this study, we investigate the distribution of the GNRs in tissue sections presenting squamous cell carcinoma (SCC) to evaluate the SCC margins. Air scanning electron microscopy (airSEM), a novel, high resolution microscopy is used, enabling to localize and actually visualize nanoparticles on the tissue. The airSEM pictures presented a gradient of GNRs from the tumor to normal epithelium, spread in an area of 1 mm, suggesting tumor margins of 1 mm. Diffusion reflection (DR) measurements, performed in a resolution of 1 mm, of human oral SCC have shown a clear difference between the DR profiles of the healthy epithelium and the tumor itself.
Collapse
Affiliation(s)
- Rinat Ankri
- Faculty of Engineering and Institute of Nanotechnology and Advanced Materials, Bar-Ilan University , Ramat-Gan 5290002, Israel
| | - Ariel Ashkenazy
- Faculty of Engineering and Institute of Nanotechnology and Advanced Materials, Bar-Ilan University , Ramat-Gan 5290002, Israel
| | | | | | - Asaf Olshinka
- Department of Plastic Surgery, Rabin Medical Center , Petach Tikva 4941492, Israel
| | - Nitza Goldenberg-Cohen
- Pediatric Unit, Ophthalmology Department, Schneider Children's Medical Center of Israel, Petach Tikva, Israel and the Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv 6997801, Israel
| | - Aron Popovtzer
- Davidoff Cancer Center, Rabin Medical Center , Beilinson Campus, Petah Tiqwa 4941492, Israel
| | - Dror Fixler
- Faculty of Engineering and Institute of Nanotechnology and Advanced Materials, Bar-Ilan University , Ramat-Gan 5290002, Israel
| | - Abraham Hirshberg
- Department of Oral Pathology and Oral Medicine, The Maurice and Gabriela Goldschleger School of Dental Medicine, Tel Aviv University , Tel Aviv 6423906, Israel
| |
Collapse
|