1
|
Kumar A, George JM, Sharma S, Koyyadi S, Sharma SK, Verwilst P, Bhatia A, Patro SK, Aggarwal A, Gupta S, Sharma S, Sharma A. pH-Activatable Molecular Probe for COX-2 Imaging in Human Oral Squamous Carcinoma Cells and Patient-Derived Tissues. ACS APPLIED BIO MATERIALS 2024; 7:8517-8527. [PMID: 39561328 DOI: 10.1021/acsabm.4c01323] [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: 11/21/2024]
Abstract
For developing a successful cancer therapeutic modality, the early precise detection of cancer cells in patient biopsies in oral squamous cell carcinoma (OSCC) is crucial. This could help researchers create new diagnostic and therapeutic tools and assist clinicians in recommending more effective treatment plans and improving patient survival. We have developed an SMPD, cyclooxygenase-2 (COX-2) targeting pH-activable fluorophore named CNP, combining a potent COX-2 inhibitor, celecoxib, linked to a naphthalimide fluorophore with an acidic microenvironment-responsive piperazine moiety for specific optical imaging of OSCC in cells and patient tissues. Compared to reference probe RNP lacking celecoxib, CNP selectively enters the COX-2 overexpressing oral cancer cells. Its acidity-responsive imaging response enhances selectivity over cancers with lower COX-2 expression levels and normal cells. Further, CNP is demonstrated in imaging OSCC cells in patient-derived biopsies. Thus, multifunctional CNP shows potential in exploring more reagents for fluorescence-based detection of OSCC cells in patient tissues with translational applications.
Collapse
Affiliation(s)
- Akhil Kumar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
- Oral Health Sciences Centre, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Jiya Mary George
- Amity School of Chemical Sciences, Amity University Punjab, Sector 82, Mohali, Punjab 140306, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
- CSIR-Central Scientific Instruments Organisation, Sector 30C, Chandigarh 160030, India
| | - Sushank Sharma
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Sundar Koyyadi
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Suchinder K Sharma
- Amity School of Physical Sciences, Amity University Punjab, Sector 82, Mohali, Punjab 140306, India
| | - Peter Verwilst
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, Box 1041, Leuven 3000, Belgium
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Sourabha Kumar Patro
- Department of Otolaryngology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Anjali Aggarwal
- Department of Anatomy, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Shipra Gupta
- Oral Health Sciences Centre, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Sheetal Sharma
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | - Amit Sharma
- Amity School of Chemical Sciences, Amity University Punjab, Sector 82, Mohali, Punjab 140306, India
| |
Collapse
|
2
|
Liu L, Yang M, Chen Z. Surface functionalized nanomaterial systems for targeted therapy of endocrine related tumors: a review of recent advancements. Drug Deliv 2024; 31:2390022. [PMID: 39138394 PMCID: PMC11328606 DOI: 10.1080/10717544.2024.2390022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
The application of multidisciplinary techniques in the management of endocrine-related cancers is crucial for harnessing the advantages of multiple disciplines and their coordinated efforts in eliminating tumors. Due to the malignant characteristics of cancer cells, they possess the capacity to develop resistance to traditional treatments such as chemotherapy and radiotherapy. Nevertheless, despite diligent endeavors to enhance the prediction of outcomes, the overall survival rate for individuals afflicted with endocrine-related malignancy remains quite miserable. Hence, it is imperative to investigate innovative therapy strategies. The latest advancements in therapeutic tactics have offered novel approaches for the therapy of various endocrine tumors. This paper examines the advancements in nano-drug delivery techniques and the utilization of nanomaterials for precise cancer cures through targeted therapy. This review provides a thorough analysis of the potential of combined drug delivery strategies in the treatment of thyroid cancer, adrenal gland tumors, and pancreatic cancer. The objective of this study is to gain a deeper understanding of current therapeutic approaches, stimulate the development of new drug DDS, and improve the effectiveness of treatment for patients with these diseases. The intracellular uptake of pharmaceuticals into cancer cells can be significantly improved through the implantation of synthetic or natural substances into nanoparticles, resulting in a substantial reduction in the development of endocrine malignancies.
Collapse
Affiliation(s)
- Limei Liu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Miao Yang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ziyang Chen
- Department of Gastroenterology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
3
|
Kerpa S, Holzapfel M, Staufer T, Kuhrwahl R, Mutas M, Werner S, Schulze VR, Nakielski P, Feliu N, Oetjen E, Haak J, Ziegler F, Buchin R, Han J, Parak WJ, Grüner F, Maison W. Iodinated PSMA Ligands as XFI Tracers for Targeted Cell Imaging and Characterization of Nanoparticles. Int J Mol Sci 2024; 25:11880. [PMID: 39595950 PMCID: PMC11594147 DOI: 10.3390/ijms252211880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/18/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Prostate cancer is the second most commonly diagnosed cancer in men worldwide. Despite this, current diagnostic tools are still not satisfactory, lacking sensitivity for early-stage or single-cell diagnosis. This study describes the development of small-molecule tracers for the well-known tumor marker prostate-specific membrane antigen (PSMA). These tracers contain a urea motif for PSMA-targeting and iodinated aromatic moieties to allow detection via X-ray fluorescence imaging (XFI). Tracers with a triiodobenzoyl moiety allowed the specific targeting and successful imaging of PSMA+ cell lines with XFI. The XFI-measured uptake of 7.88 × 10-18 mol iodine (I) per cell is consistent with the uptake of known PSMA tracers measured by other techniques such as inductively coupled plasma mass spectrometry (ICP-MS). This is the first successful application of XFI to tumor cell targeting with a small-molecule tracer. In addition, iodinated tracers were used for the characterization of quantum dots (QDs) conjugated to PSMA-targeting urea motifs. The resulting targeted QD conjugates were shown to selectively bind PSMA+ cell lines via confocal microscopy. The immobilized iodinated targeting vectors allowed the determination of the tracer/QD ratio via XFI and ICP-MS. This ratio is a key property of targeted particles and difficult to measure by other techniques.
Collapse
Affiliation(s)
- Svenja Kerpa
- Department of Chemistry, Institute of Pharmacy, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany;
| | - Malte Holzapfel
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
| | - Theresa Staufer
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Robert Kuhrwahl
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Marina Mutas
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany;
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany;
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Verena R. Schulze
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
| | - Pascal Nakielski
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
| | - Neus Feliu
- Fraunhofer Institute for Applied Polymer Research IAP, Center for Applied Nanotechnology CAN, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany; (M.H.); (M.M.); (V.R.S.); (P.N.); (N.F.)
| | - Elke Oetjen
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany;
| | - Jannis Haak
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Florian Ziegler
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Rasmus Buchin
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Jili Han
- Department of Physics, Universität Hamburg and Center for Hybrid Nanostructures (CHyN), Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (W.J.P.)
| | - Wolfgang J. Parak
- Department of Physics, Universität Hamburg and Center for Hybrid Nanostructures (CHyN), Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (W.J.P.)
| | - Florian Grüner
- University of Hamburg and Center for Free-Electron Laser Science, Luruper Chaussee 149, 22761 Hamburg, Germany; (T.S.); (R.K.); (J.H.); (F.Z.); (R.B.)
| | - Wolfgang Maison
- Department of Chemistry, Institute of Pharmacy, Universität Hamburg, Bundesstrasse 45, 20146 Hamburg, Germany;
| |
Collapse
|
4
|
Oblak ML, Lu HY, Ram AS, McKenna C. Comparative aspects of targeted sentinel lymph node mapping in veterinary and human medicine: opportunities for future research. Front Med (Lausanne) 2024; 11:1342456. [PMID: 38633313 PMCID: PMC11021648 DOI: 10.3389/fmed.2024.1342456] [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: 11/21/2023] [Accepted: 02/29/2024] [Indexed: 04/19/2024] Open
Abstract
There is a significant overlap in the genetic, metabolic and epigenetic alterations between human and companion animal cancers, including those of the oral cavity, breast, bladder, skin, lungs and pancreas. In many cancer types, the identification and removal of affected lymph nodes are essential for accurate cancer management, including treatment and prognosis. Historically, lymphadenectomy and subsequent radical resection based on regional anatomy, palpation and lymph node aspirates were considered sufficient; however, modern approaches with sentinel lymph node mapping (SLN) mapping have increased the accuracy of surgical decision-making. Preoperative and intraoperative SLN mapping techniques in veterinary patients parallel those used in human medicine. While many of these techniques are highly successful, the main challenges with current methodologies are their sensitivity and specificity for the presence of cancer, which can be overcome via precision medicine and targeted SLN mapping agents. Given the large population of dogs and cats with cancer, the crossover of knowledge between species can help to deepen our understanding of many of these cancers and can be useful in evaluating new drugs and/or therapies. In this review, we discuss SLN mapping techniques in veterinary medicine and the concept of precision medicine as it relates to targeted SLN mapping imaging agents. The large number of companion animals affected by cancer is an underutilized resource to bridge the translational gap and we aim to provide a reference for the use of dogs and cats as a comparative model for human SLN mapping.
Collapse
Affiliation(s)
- Michelle L. Oblak
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Hui Yu Lu
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Ann S. Ram
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Charly McKenna
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
5
|
Kravchenko Y, Sikora K, Wireko AA, Lyndin M. Fluorescence visualization for cancer DETECTION: EXPERIENCE and perspectives. Heliyon 2024; 10:e24390. [PMID: 38293525 PMCID: PMC10827512 DOI: 10.1016/j.heliyon.2024.e24390] [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: 01/11/2023] [Revised: 12/24/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
The current review focuses on the latest advances in the improvement and application of fluorescence imaging technology. Near-infrared (NIR) fluorescence imaging is a promising new technique that uses non-specific fluorescent agents and targeted fluorescent tracers combined with a dedicated camera to better navigate and visualize tumors. Fluorescence-guided surgery (FGS) is used to perform various tasks, helping the surgeon to distinguish lymphatic vessels and nodes from surrounding tissues easily and quickly assess the perfusion of the planned resection area, including intraoperative visualization of metastases. The results of the insertion of fluorescence visualization as an auxiliary method to cancer detection and high-risk metastatic lesions in clinical practice have demonstrated enthusiastic results and huge potential. However, intraoperative fluorescence visualization must not be considered as a main diagnostic or treatment method but as an aid to the surgeon. Thus, fluorescence study does not dispense the diagnostic gold standards of benign or malignant tumors (conventional examination, biopsy, ultrasonography and computed tomography, etc.) and can be done usually during intraoperative treatment. Moreover, as fluorescence surgery and fluorescence diagnostic techniques continue to improve, it is likely that they will evolve towards targeted fluorescence imaging probes that will increasingly target a specific type of cancer cell. The most important point remains the search for highly selective messengers of fluorescent labels, which make it possible to identify tumor cells exclusively in the affected organs and indicate to surgeons the boundaries of their spread and metastasis.
Collapse
Affiliation(s)
- Yaroslav Kravchenko
- Sumy State University, Sumy, Ukraine
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Poznan, Poland
| | | | | | - Mykola Lyndin
- Sumy State University, Sumy, Ukraine
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Essen, 45147, Germany
| |
Collapse
|
6
|
Maschio R, Buonsanti F, Crivellin F, Ferretti F, Lattuada L, Maisano F, Orio L, Pizzuto L, Campanella R, Clouet A, Cavallotti C, Giovenzana GB. Improved synthesis of DA364, an NIR fluorescence RGD probe targeting α vβ 3 integrin. Org Biomol Chem 2023; 21:8584-8592. [PMID: 37855098 DOI: 10.1039/d3ob01206a] [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: 10/20/2023]
Abstract
Optical imaging (OI) is gaining increasing attention in medicine as a non-invasive diagnostic imaging technology and as a useful tool for image-guided surgery. OI exploits the light emitted in the near-infrared region by fluorescent molecules able to penetrate living tissues. Cyanines are an important class of fluorescent molecules and by their conjugation to peptides it is possible to achieve optical imaging of tumours by selective targeting. We report here the improvements obtained in the synthesis of DA364, a small fluorescent probe (1.5 kDa) prepared by conjugation of pentamethine cyanine Cy5.5 to an RGD peptidomimetic, which can target tumour cells overexpressing integrin αvβ3 receptors.
Collapse
Affiliation(s)
- Rachele Maschio
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Largo Donegani 2/3, 28100 Novara, Italy.
| | - Federica Buonsanti
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Federico Crivellin
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Fulvio Ferretti
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Luciano Lattuada
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Federico Maisano
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Laura Orio
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Lorena Pizzuto
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010 Colleretto Giacosa, TO, Italy.
| | - Raphael Campanella
- Bracco Suisse SA, Route de la Galaise 31, 1228 Plan le Ouates, Switzerland
| | - Anthony Clouet
- Bracco Suisse SA, Route de la Galaise 31, 1228 Plan le Ouates, Switzerland
| | | | - Giovanni B Giovenzana
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Largo Donegani 2/3, 28100 Novara, Italy.
- CAGE Chemicals Srl, Via Bovio 6, 28100 Novara, Italy
| |
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: 0] [Impact Index Per Article: 0] [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
|
Kwon MJ, House BJ, Barth CW, Solanki A, Jones JA, Davis SC, Gibbs SL. Dual probe difference specimen imaging for prostate cancer margin assessment. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:082806. [PMID: 37082104 PMCID: PMC10111791 DOI: 10.1117/1.jbo.28.8.082806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 03/31/2023] [Indexed: 05/03/2023]
Abstract
Significance Positive margin status due to incomplete removal of tumor tissue during radical prostatectomy for high-risk localized prostate cancer requires reoperation or adjuvant therapy, which increases morbidity and mortality. Adverse effects of prostate cancer treatments commonly include erectile dysfunction, urinary incontinence, and bowel dysfunction, making successful initial curative prostatectomy imperative. Aim Current intraoperative tumor margin assessment is largely limited to frozen section analysis, which is a lengthy, labor-intensive process that is obtrusive to the clinical workflow within the operating room (OR). Therefore, a rapid method for prostate cancer margin assessment in the OR could improve outcomes for patients. Approach Dual probe difference specimen imaging (DDSI), which uses paired antibody-based probes that are labeled with spectrally distinct fluorophores, was shown herein for prostate cancer margin assessment. The paired antibody-based probes consisted of a targeted probe to prostate-specific membrane antigen (PSMA) and an untargeted probe, which were used as a cocktail to stain resected murine tissue specimens including prostate tumor, adipose, muscle, and normal prostate. Ratiometric images (i.e., DDSI) of the difference between targeted and untargeted probe uptake were calculated and evaluated for accuracy using receiver operator characteristic curve analysis with area under the curve values used to evaluate the utility of the DDSI method to detect PSMA positive prostate cancer. Results Targeted and untargeted probe uptake was similar between the high and low PSMA expressing tumor due to nonspecific probe uptake after topical administration. The ratiometric DDSI approach showed substantial contrast difference between the PSMA positive tumors and their respective normal tissues (prostate, adipose, muscle). Furthermore, DDSI showed substantial contrast difference between the high PSMA expressing tumors and the minimally PSMA expressing tumors due to the ratiometric correction for the nonspecific uptake patterns in resected tissues. Conclusions Previous work has shown that ratiometic imaging has strong predictive value for breast cancer margin status using topical administration. Translation of the ratiometric DDSI methodology herein from breast to prostate cancers demonstrates it as a robust, ratiometric technique that provides a molecularly specific imaging modality for intraoperative margin detection. Using the validated DDSI protocol on resected prostate cancers permitted rapid and accurate assessment of PSMA status as a surrogate for prostate cancer margin status. Future studies will further evaluate the utility of this technology to quantitatively characterize prostate margin status using PSMA as a biomarker.
Collapse
Affiliation(s)
- Marcus J. Kwon
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Broderick J. House
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Connor W. Barth
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Allison Solanki
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Jocelyn A. Jones
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
| | - Scott C. Davis
- Thayer School of Engineering at Dartmouth College, Hanover, New Hampshire, United States
| | - Summer L. Gibbs
- Oregon Health & Science University, Biomedical Engineering Department, Portland, Oregon, United States
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States
- Address all correspondence to Summer L. Gibbs,
| |
Collapse
|
9
|
Lu G, Han Z, Hu M. Optical imaging technology realizes early tumor diagnosis by detecting angiogenesis. Microsc Res Tech 2023; 86:232-241. [PMID: 36412215 DOI: 10.1002/jemt.24262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 10/25/2022] [Accepted: 11/06/2022] [Indexed: 11/23/2022]
Abstract
The occurrence and development of blood vessels play a key role in different stages of tumor growth, while current imaging techniques are difficult to detect early tumor angiogenesis because of their low sensitivity. Therefore, this article introduces high-sensitivity optical imaging technology to achieve early tumor diagnosis by detecting tumor angiogenesis. Liver and pancreatic tumor models in nude mice were respectively established to represent tumors with a rich or poor blood supply. The two optical imaging methods, in vivo confocal fluorescence imaging and photoacoustic imaging, were used to detect tumor angiogenesis at different stages. Finally, the changes in blood vessels were verified by immunostaining. Both autoluminescence imaging and pathological staining confirmed that these two tumor models were successfully established. In vivo confocal fluorescence imaging found that the early tumor blood vessel structure had obvious characteristics: disorder, tortuous deformation, thin diameter, which were significantly different from the normal tissues. Photoacoustic imaging could effectively identify blood vessels inside early tumors, which were small and disordered and might be used as one of the predictors of early tumor development. CD31 immunostaining was used to evaluate the vascular status of tumors at different stages and under different blood supply conditions. The vascular structures observed under the microscope in the two tumor models were consistent with the results observed by optical imaging methods. The optical imaging methods could monitor the characteristics of angiogenesis in the rich or poor blood supply tumors, especially the early diagnosis of tumors.
Collapse
Affiliation(s)
- Guanhua Lu
- Department of Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ziyu Han
- Department of Ultrasonic Diagnosis, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Min Hu
- Department of Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
10
|
García de Jalón E, Kleinmanns K, Fosse V, Davidson B, Bjørge L, Haug BE, McCormack E. Comparison of Five Near-Infrared Fluorescent Folate Conjugates in an Ovarian Cancer Model. Mol Imaging Biol 2023; 25:144-155. [PMID: 34888759 PMCID: PMC9971101 DOI: 10.1007/s11307-021-01685-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Fluorescence imaging (FLI) using targeted near-infrared (NIR) conjugates aids the detection of tumour lesions pre- and intraoperatively. The optimisation of tumour visualisation and contrast is essential and can be achieved through high tumour-specificity and low background signal. However, the choice of fluorophore is recognised to alter biodistribution and clearance of conjugates and is therefore a determining factor in the specificity of target binding. Although ZW800-1, IRDye® 800CW and ICG are the most commonly employed NIR fluorophores in clinical settings, the fluorophore with optimal in vivo characteristics has yet to be determined. Therefore, we aimed to characterise the impact the choice of fluorophore has on the biodistribution, specificity and contrast, by comparing five different NIR fluorophores conjugated to folate, in an ovarian cancer model. PROCEDURES ZW800-1, ZW800-1 Forte, IRDye® 800CW, ICG-OSu and an in-house synthesised Cy7 derivative were conjugated to folate through an ethylenediamine linker resulting in conjugates 1-5, respectively. The optical properties of all conjugates were determined by spectroscopy, the specificity was assessed in vitro by flow cytometry and FLI, and the biodistribution was studied in vivo and ex vivo in a subcutaneous Skov-3 ovarian cancer model. RESULTS We demonstrated time- and receptor-dependent binding of folate conjugates in vitro and in vivo. Healthy tissue clearance characteristics and tumour-specific signal varied between conjugates 1-5. ZW800-1 Forte (2) revealed the highest contrast in folate receptor alpha (FRα)-positive xenografts and showed statistically significant target specificity. While conjugates 1, 2 and 3 are renally cleared, hepatobiliary excretion and no or very low accumulation in tumours was observed for 4 and 5. CONCLUSIONS The choice of fluorophore has a significant impact on the biodistribution and tumour contrast. ZW800-1 Forte (2) exhibited the best properties of those tested, with significant specific fluorescence signal.
Collapse
Affiliation(s)
- Elvira García de Jalón
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021, Bergen, Norway.,Department of Chemistry and Centre for Pharmacy, University of Bergen, Allégaten 41, N-5007, Bergen, Norway
| | - Katrin Kleinmanns
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021, Bergen, Norway
| | - Vibeke Fosse
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021, Bergen, Norway
| | - Ben Davidson
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, and Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Line Bjørge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021, Bergen, Norway.,Department of Obstetrics and Gynaecology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Bengt Erik Haug
- Department of Chemistry and Centre for Pharmacy, University of Bergen, Allégaten 41, N-5007, Bergen, Norway.
| | - Emmet McCormack
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021, Bergen, Norway. .,Centre for Pharmacy, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021, Bergen, Norway. .,Vivarium, Department of Clinical Science, The University of Bergen, Jonas Lies vei 65, 5021, Bergen, Norway.
| |
Collapse
|
11
|
Image-guided drug delivery in nanosystem-based cancer therapies. Adv Drug Deliv Rev 2023; 192:114621. [PMID: 36402247 DOI: 10.1016/j.addr.2022.114621] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/18/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
The past decades have shown significant advancements in the development of solid tumor treatment. For instance, implementation of nanosystems for drug delivery has led to a reduction in side effects and improved delivery to the tumor region. However, clinical translation has faced challenges, as tumor drug levels are still considered to be inadequate. Interdisciplinary research has resulted in the development of more advanced drug delivery systems. These are coined "smart" due to the ability to be followed and actively manipulated in order to have better control over local drug release. Therefore, image-guided drug delivery can be a powerful strategy to improve drug activity at the target site. Being able to visualize the inflow of the administered smart nanosystem within the tumor gives the potential to determine the right moment to apply the facilitator to initiate drug release. Here we provide an overview of available nanosystems, imaging moieties, and imaging techniques. We discuss preclinical application of these smart drug delivery systems, the strength of image-guided drug delivery, and the future of personalized treatment.
Collapse
|
12
|
Ma H, Jiang K, Hong Y, Lei Y, Fan Y, Jiang W, Zhao L, Liu J, Yao W, Xu J, He M, Wei M. Screening of an annexin-A2-targeted heptapeptide for pancreatic adenocarcinoma localization. Mol Oncol 2022; 17:872-886. [PMID: 36453020 PMCID: PMC10158761 DOI: 10.1002/1878-0261.13352] [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: 08/11/2022] [Revised: 11/04/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
Annexin A2 (ANXA2) encodes an oncoprotein whose expression has been found to correlate with poorer overall survival (OS) of pancreatic adenocarcinoma (PAAD) patients. Although peptides are available for targeting ANXA2, none of these were initially selected to target this protein specifically. Here, we took ANXA2 as a molecular target for PAAD and employed the phage display technique to screen for a new ANXA2-targeted peptide. The resultant heptapeptide, YW7, was firstly labeled with fluorescein isothiocyanate (FITC) to evaluate its selectivity in cellular uptake, and further with the near-infrared fluorescent (NIRF) dye Cy7 to assess in vivo distribution in a mouse model bearing PANC-1 human pancreatic cancer xenografic tumors. We found that both FITC-YW7 and Cy7-YW7 probes showed significantly higher uptake in PANC-1 cells compared to the HPDE6-C7 pancreatic epithelium cells. Mice intravenously injected with Cy7-YW7 showed higher tumor-to-background ratios (TBRs) (~ 2.7-fold) in tumor tissues compared to those injected with Cy7 alone. Our study suggested that YW7 is a novel peptide targeting ANXA2 and Cy7-YW7 is an NIRF probe potentially useful for the early detection of PAAD.
Collapse
Affiliation(s)
- Heyao Ma
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Kai Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yuhan Hong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yu Lei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yue Fan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Wenjian Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lei Zhao
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of China Medical University, Shenyang, China
| | - Jinyang Liu
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of China Medical University, Shenyang, China
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Jiao Xu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China.,Liaoning Medical Diagnosis and Treatment Center, Shenyang, China
| |
Collapse
|
13
|
Azari F, Zhang K, Kennedy GT, Chang A, Nadeem B, Delikatny EJ, Singhal S. Precision Surgery Guided by Intraoperative Molecular Imaging. J Nucl Med 2022; 63:1620-1627. [PMID: 35953303 PMCID: PMC9635678 DOI: 10.2967/jnumed.121.263409] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Intraoperative molecular imaging (IMI) has recently emerged as an important tool in the armamentarium of surgical oncologists. IMI allows real-time assessment of oncologic resection quality, margin assessment, and occult disease detection during real-time surgery. Numerous tracers have now been developed for use in IMI-guided tissue sampling. Fluorochromes localize to the tumor by taking advantage of their disorganized capillary milieu, overexpressed receptors, or upregulated enzymes. Although fluorescent tracers can suffer from issues of autofluorescence and lack of depth penetration, these challenges are being addressed through hybrid radioactive/fluorescent tracers and new tracers that fluoresce in the near-infrared (NIR-II [wavelength > 1,000 nm]) range. IMI is already being used to treat numerous cancers, with demonstrated improvement in cancer recurrence and patient outcomes without incurring significant burden on either clinicians or patients. In this comprehensive review, we discuss history, mechanism, current oncologic applications, and future directions of IMI-guided optical biopsy.
Collapse
Affiliation(s)
- Feredun Azari
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kevin Zhang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Gregory T. Kennedy
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ashley Chang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Bilal Nadeem
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania;
| |
Collapse
|
14
|
Fe3O4/Graphene-Based Nanotheranostics for Bimodal Magnetic Resonance/Fluorescence Imaging and Cancer Therapy. J Inorg Organomet Polym Mater 2022. [DOI: 10.1007/s10904-022-02457-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
15
|
A comprehensive review on different approaches for tumor targeting using nanocarriers and recent developments with special focus on multifunctional approaches. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00583-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
16
|
López-Álvarez M, Heuker M, Sjollema KA, van Dam GM, van Dijl JM, IJpma FFA, van Oosten M. Bacteria-targeted fluorescence imaging of extracted osteosynthesis devices for rapid visualization of fracture-related infections. Eur J Nucl Med Mol Imaging 2022; 49:2276-2289. [PMID: 35079847 PMCID: PMC9165280 DOI: 10.1007/s00259-022-05695-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/19/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Fracture-related infection (FRI) is a serious complication in orthopedic trauma surgery worldwide. Especially, the distinction of infection from sterile inflammation and the detection of low-grade infection are highly challenging. The objective of the present study was to obtain proof-of-principle for the use of bacteria-targeted fluorescence imaging to detect FRI on extracted osteosynthesis devices as a step-up towards real-time image-guided trauma surgery. METHODS Extracted osteosynthesis devices from 13 patients, who needed revision surgery after fracture treatment, were incubated with a near-infrared fluorescent tracer composed of the antibiotic vancomycin and the fluorophore IRDye800CW (i.e., vanco-800CW). Subsequently, the devices were imaged, and vanco-800CW fluorescence signals were correlated to the results of microbiological culturing and to bacterial growth upon replica plating of the imaged devices on blood agar. RESULTS Importantly, compared to culturing, the bacteria-targeted fluorescence imaging of extracted osteosynthesis devices with vanco-800CW allows for a prompt diagnosis of FRI, reducing the time-to-result from days to less than 30 min. Moreover, bacteria-targeted imaging can provide surgeons with real-time visual information on the presence and extent of infection. CONCLUSION Here, we present the first clinical application of fluorescence imaging for the detection of FRI. We conclude that imaging with vanco-800CW can provide early, accurate, and real-time visual diagnostic information on FRI in the clinical setting, even in the case of low-grade infections.
Collapse
Affiliation(s)
- Marina López-Álvarez
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO BOX 30001, 9700 RB, Groningen, The Netherlands
| | - Marjolein Heuker
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO BOX 30001, 9700 RB, Groningen, The Netherlands
| | - Klaas A Sjollema
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gooitzen M van Dam
- Departments of Surgery, Nuclear Medicine and Molecular Imaging, Medical Imaging Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- TRACER Europe B.V./AxelaRx, Groningen, The Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO BOX 30001, 9700 RB, Groningen, The Netherlands.
| | - Frank F A IJpma
- Department of Surgery, Division of Trauma Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marleen van Oosten
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO BOX 30001, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
17
|
Lauwerends LJ, Abbasi H, Bakker Schut TC, Van Driel PBAA, Hardillo JAU, Santos IP, Barroso EM, Koljenović S, Vahrmeijer AL, Baatenburg de Jong RJ, Puppels GJ, Keereweer S. The complementary value of intraoperative fluorescence imaging and Raman spectroscopy for cancer surgery: combining the incompatibles. Eur J Nucl Med Mol Imaging 2022; 49:2364-2376. [PMID: 35102436 PMCID: PMC9165240 DOI: 10.1007/s00259-022-05705-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/23/2022] [Indexed: 01/09/2023]
Abstract
A clear margin is an important prognostic factor for most solid tumours treated by surgery. Intraoperative fluorescence imaging using exogenous tumour-specific fluorescent agents has shown particular benefit in improving complete resection of tumour tissue. However, signal processing for fluorescence imaging is complex, and fluorescence signal intensity does not always perfectly correlate with tumour location. Raman spectroscopy has the capacity to accurately differentiate between malignant and healthy tissue based on their molecular composition. In Raman spectroscopy, specificity is uniquely high, but signal intensity is weak and Raman measurements are mainly performed in a point-wise manner on microscopic tissue volumes, making whole-field assessment temporally unfeasible. In this review, we describe the state-of-the-art of both optical techniques, paying special attention to the combined intraoperative application of fluorescence imaging and Raman spectroscopy in current clinical research. We demonstrate how these techniques are complementary and address the technical challenges that have traditionally led them to be considered mutually exclusive for clinical implementation. Finally, we present a novel strategy that exploits the optimal characteristics of both modalities to facilitate resection with clear surgical margins.
Collapse
Affiliation(s)
- L J Lauwerends
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - H Abbasi
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
- Center for Optical Diagnostics and Therapy, Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - T C Bakker Schut
- Center for Optical Diagnostics and Therapy, Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - P B A A Van Driel
- Department of Orthopedic Surgery, Isala Hospital, Zwolle, Netherlands
| | - J A U Hardillo
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - I P Santos
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| | | | - S Koljenović
- Department of Pathology, Antwerp University Hospital/Antwerp University, Antwerp, Belgium
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - R J Baatenburg de Jong
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - G J Puppels
- Center for Optical Diagnostics and Therapy, Department of Dermatology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - S Keereweer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands.
| |
Collapse
|
18
|
Monaco H, Yokomizo S, Choi HS, Kashiwagi S. Quickly evolving near‐infrared photoimmunotherapy provides multifaceted approach to modern cancer treatment. VIEW 2022. [DOI: 10.1002/viw.20200110] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Hailey Monaco
- Gordon Center for Medical Imaging Department of Radiology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts USA
| | - Shinya Yokomizo
- Gordon Center for Medical Imaging Department of Radiology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts USA
- Department of Radiological Sciences Tokyo Metropolitan University Arakawa Tokyo Japan
| | - Hak Soo Choi
- Gordon Center for Medical Imaging Department of Radiology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging Department of Radiology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts USA
| |
Collapse
|
19
|
Thapaliya ER, Usama SM, Patel NL, Feng Y, Kalen JD, St Croix B, Schnermann MJ. Cyanine Masking: A Strategy to Test Functional Group Effects on Antibody Conjugate Targeting. Bioconjug Chem 2022; 33:718-725. [PMID: 35389618 PMCID: PMC10506421 DOI: 10.1021/acs.bioconjchem.2c00083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Conjugates of small molecules and antibodies are broadly employed diagnostic and therapeutic agents. Appending a small molecule to an antibody often significantly impacts the properties of the resulting conjugate. Here, we detail a systematic study investigating the effect of various functional groups on the properties of antibody-fluorophore conjugates. This was done through the preparation and analysis of a series of masked heptamethine cyanines (CyMasks)-bearing amides with varied functional groups. These were designed to exhibit a broad range of physical properties, and include hydrophobic (-NMe2), pegylated (NH-PEG-8 or NH-PEG-24), cationic (NH-(CH2)2NMe3+), anionic (NH-(CH2)2SO3-), and zwitterionic (N-(CH2)2NMe3+)-(CH2)3SO3-) variants. The CyMask series was appended to monoclonal antibodies (mAbs) and analyzed for the effects on tumor targeting, clearance, and non-specific organ uptake. Among the series, zwitterionic and pegylated dye conjugates had the highest tumor-to-background ratio (TBR) and a low liver-to-background ratio. By contrast, the cationic and zwitterionic probes had high tumor signal and high TBR, although the latter also exhibited an elevated liver-to-background ratio (LBR). Overall, these studies provide a strategy to test the functional group effects and suggest that zwitterionic substituents possess an optimal combination of high tumor signal, TBR, and low LBR. These results suggest an appealing strategy to mask hydrophobic payloads, with the potential to improve the properties of bioconjugates in vivo.
Collapse
Affiliation(s)
- Ek Raj Thapaliya
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Nimit L Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute, NIH, Frederick, Maryland 21702, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute, NIH, Frederick, Maryland 21702, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
20
|
Fukuda T, Yokomizo S, Casa S, Monaco H, Manganiello S, Wang H, Lv X, Ulumben AD, Yang C, Kang MW, Inoue K, Fukushi M, Sumi T, Wang C, Kang H, Bao K, Henary M, Kashiwagi S, Soo Choi H. Fast and Durable Intraoperative Near-infrared Imaging of Ovarian Cancer Using Ultrabright Squaraine Fluorophores. Angew Chem Int Ed Engl 2022; 61:e202117330. [PMID: 35150468 PMCID: PMC9007913 DOI: 10.1002/anie.202117330] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Indexed: 12/19/2022]
Abstract
The residual tumor after surgery is the most significant prognostic factor of patients with epithelial ovarian cancer. Near-infrared (NIR) fluorescence-guided surgery is actively utilized for tumor localization and complete resection during surgery. However, currently available contrast-enhancing agents display low on-target binding, unfavorable pharmacokinetics, and toxicity, thus not ideal for clinical use. Here we report ultrabright and stable squaraine fluorophores with optimal pharmacokinetics by introducing an asymmetric molecular conformation and surface charges for rapid transporter-mediated cellular uptake. Among the tested, OCTL14 shows low serum binding and rapid distribution into cancer tissue via organic cation transporters (OCTs). Additionally, the charged squaraine fluorophores are retained in lysosomes, providing durable intraoperative imaging in a preclinical murine model of ovarian cancer up to 24 h post-injection. OCTL14 represents a significant departure from the current bioconjugation approach of using a non-targeted fluorophore and would provide surgeons with an indispensable tool to achieve optimal resection.
Collapse
Affiliation(s)
- Takeshi Fukuda
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, 1-4-3, Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shinya Yokomizo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo, 116-8551, Japan
| | - Stefanie Casa
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Hailey Monaco
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Sophia Manganiello
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Haoran Wang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Xiangmin Lv
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amy Daniel Ulumben
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Chengeng Yang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Min-Woong Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Chungnam National University, Daejeon, 301-721, South Korea
| | - Kazumasa Inoue
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo, 116-8551, Japan
| | - Masahiro Fukushi
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa, Tokyo, 116-8551, Japan
| | - Toshiyuki Sumi
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, 1-4-3, Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kai Bao
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Maged Henary
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
- Center for Diagnostics and Therapeutics, 145 Piedmont Avenue S.E., Atlanta, GA 30303, USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
21
|
Dell’Oglio P, Mazzone E, Buckle T, Maurer T, Navab N, van Oosterom MN, Schilling C, Witjes MJH, Vahrmeijer AL, Klode J, Vojnovic B, Mottrie A, van der Poel HG, Hamdy F, van Leeuwen FWB. Precision surgery: the role of intra-operative real-time image guidance - outcomes from a multidisciplinary European consensus conference. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2022; 12:74-80. [PMID: 35535122 PMCID: PMC9077167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
Developments within the field of image-guided surgery are ever expanding, driven by collective involvement of clinicians, researchers, and industry. While the general conception of the potential of image-guided surgery is to improve surgical outcome, the specific motives and goals that drive can differ between the different expert groups. To establish the current and future role of intra-operative image guidance within the field of image-guided surgery a Delphi consensus survey was conducted during the 2nd European Congress on Image-guided surgery. This multidisciplinary survey included questions on the conceptual potential and clinical value of image-guided surgery and was aimed at defining specific areas of research and development in the field in order to stimulate further advances towards precision surgery. Obtained results based on questionnaires filled in by 56 panel experts (clinicians: N=30, researchers: N=20 and industry: N=6) were discussed during a dedicated expert discussion session during the conference. The outcome of this Delphi consensus is indicative of the potential improvements offered by image-guided surgery and of the need for further research in this emerging field, that can be enriched by the identification of reliable molecular targets.
Collapse
Affiliation(s)
- Paolo Dell’Oglio
- Department of Urology, ASST Grande Ospedale Metropolitano NiguardaMilan, Italy
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek HospitalAmsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical CenterLeiden, The Netherlands
| | - Elio Mazzone
- Department of Urology and Division of Experimental Oncology, URI, Urological Research Institute, IRCCS San Raffaele Scientific InstituteMilan, Italy
| | - Tessa Buckle
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek HospitalAmsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical CenterLeiden, The Netherlands
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-EppendorfHamburg, Germany
| | - Nassir Navab
- Computer Aided Medical Procedure, Technical University of MunichMunich, Germany
| | - Matthias N van Oosterom
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek HospitalAmsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical CenterLeiden, The Netherlands
| | - Clare Schilling
- Department of Head and Neck Surgery, University College HospitalLondon, United Kingdom
| | - Max JH Witjes
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center GroningenGroningen, The Netherlands
| | | | - Joachim Klode
- Department of Dermatology, Venerology and Allergology, University Hospital Essen, University of Duisburg-EssenEssen, Germany
| | - Boris Vojnovic
- Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, University of OxfordOxford, United Kingdom
| | - Alexandre Mottrie
- ORSI, AcademyMelle, Belgium
- Department of Urology, Onze Lieve Vrouw HospitalAalst, Belgium
| | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek HospitalAmsterdam, The Netherlands
| | - Freddie Hamdy
- Nuffield Department of Surgical Sciences, University of OxfordOxford, United Kingdom
| | - Fijs WB van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek HospitalAmsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical CenterLeiden, The Netherlands
| |
Collapse
|
22
|
Bishop KW, Maitland KC, Rajadhyaksha M, Liu JTC. In vivo microscopy as an adjunctive tool to guide detection, diagnosis, and treatment. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-220032-PER. [PMID: 35478042 PMCID: PMC9043840 DOI: 10.1117/1.jbo.27.4.040601] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/05/2022] [Indexed: 05/05/2023]
Abstract
SIGNIFICANCE There have been numerous academic and commercial efforts to develop high-resolution in vivo microscopes for a variety of clinical use cases, including early disease detection and surgical guidance. While many high-profile studies, commercialized products, and publications have resulted from these efforts, mainstream clinical adoption has been relatively slow other than for a few clinical applications (e.g., dermatology). AIM Here, our goals are threefold: (1) to introduce and motivate the need for in vivo microscopy (IVM) as an adjunctive tool for clinical detection, diagnosis, and treatment, (2) to discuss the key translational challenges facing the field, and (3) to propose best practices and recommendations to facilitate clinical adoption. APPROACH We will provide concrete examples from various clinical domains, such as dermatology, oral/gastrointestinal oncology, and neurosurgery, to reinforce our observations and recommendations. RESULTS While the incremental improvement and optimization of IVM technologies should and will continue to occur, future translational efforts would benefit from the following: (1) integrating clinical and industry partners upfront to define and maintain a compelling value proposition, (2) identifying multimodal/multiscale imaging workflows, which are necessary for success in most clinical scenarios, and (3) developing effective artificial intelligence tools for clinical decision support, tempered by a realization that complete adoption of such tools will be slow. CONCLUSIONS The convergence of imaging modalities, academic-industry-clinician partnerships, and new computational capabilities has the potential to catalyze rapid progress and adoption of IVM in the next few decades.
Collapse
Affiliation(s)
- Kevin W. Bishop
- University of Washington, Department of Bioengineering, Seattle, Washington, United States
- University of Washington, Department of Mechanical Engineering, Seattle, Washington, United States
| | - Kristen C. Maitland
- Texas A&M University, Department of Biomedical Engineering, College Station, Texas, United States
| | - Milind Rajadhyaksha
- Memorial Sloan Kettering Cancer Center, Dermatology Service, New York, New York, United States
| | - Jonathan T. C. Liu
- University of Washington, Department of Bioengineering, Seattle, Washington, United States
- University of Washington, Department of Mechanical Engineering, Seattle, Washington, United States
- University of Washington, Department of Laboratory Medicine and Pathology, Seattle, Washington, United States
- Address all correspondence to Jonathan T.C. Liu,
| |
Collapse
|
23
|
Fukuda T, Yokomizo S, Casa S, Monaco H, Manganiello S, Wang H, Lv X, Ulumben AD, Yang C, Kang MW, Inoue K, Fukushi M, Sumi T, Wang C, Kang H, Bao K, Henary M, Kashiwagi S, Choi HS. Fast and Durable Intraoperative Near‐infrared Imaging of Ovarian Cancer Using Ultrabright Squaraine Fluorophores. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202117330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | | | | | - Hailey Monaco
- Massachusetts General Hospital radiology UNITED STATES
| | | | - Haoran Wang
- Massachusetts General Hospital radiology UNITED STATES
| | - Xiangmin Lv
- Massachusetts General Hospital Obstetrics and Gynecology UNITED STATES
| | | | - Chengeng Yang
- Massachusetts General Hospital radiology UNITED STATES
| | | | - Kazumasa Inoue
- Tokyo Metropolitan University - Arakawa Campus: Tokyo Toritsu Daigaku - Arakawa Campus Radiation Science JAPAN
| | - Masahiro Fukushi
- Tokyo Metropolitan University - Arakawa Campus: Tokyo Toritsu Daigaku - Arakawa Campus Radiation Science JAPAN
| | - Toshiyuki Sumi
- Osaka City University: Osaka Shiritsu Daigaku Obstetrics and Gynecology JAPAN
| | - Cheng Wang
- Massachusetts General Hospital Obstetrics and Gynecology UNITED STATES
| | - Homan Kang
- Massachusetts General Hospital radiology UNITED STATES
| | - Kai Bao
- Massachusetts General Hospital radiology UNITED STATES
| | - Maged Henary
- Georgia State University Chemistry UNITED STATES
| | - Satoshi Kashiwagi
- Massachusetts General Hospital Radiology 149 13th Street 02129 Charlestown UNITED STATES
| | - Hak Soo Choi
- Massachusetts General Hospital Radiology 149 13th Street 02129 Boston UNITED STATES
| |
Collapse
|
24
|
Michie MS, Xu B, Sudlow G, Springer LE, Pham CT, Achilefu S. Side-chain modification of collagen-targeting peptide prevents dye aggregation for improved molecular imaging of arthritic joints. J Photochem Photobiol A Chem 2022; 424:113624. [PMID: 36406204 PMCID: PMC9673490 DOI: 10.1016/j.jphotochem.2021.113624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Near-infrared (NIR) dye-peptide conjugates are widely used for tissue-targeted molecular fluorescence imaging of pathophysiologic conditions. However, the significant contribution of both dye and peptide to the net mass of these bioconjugates implies that small changes in either component could alter their photophysical and biological properties. Here, we synthesized and conjugated a type I collagen targeted peptide, RRANAALKAGELYKCILY, to either a hydrophobic (LS1000) or hydrophilic (LS1006) NIR fluorescent dye. Spectroscopic analysis revealed rapid self-assembly of both LS1000 and LS1006 in aqueous media to form stable dimeric/H aggregates, regardless of the free dye's solubility in water. We discovered that replacing the cysteine residue in LS1000 and LS1006 with acetamidomethyl cysteine to afford LS1001 and LS1107, respectively, disrupted the peptide's self-assembly and activated the previously quenched dye's fluorescence in aqueous conditions. These results highlight the dominant role of the octadecapeptide, but not the dye molecules, in controlling the photophysical properties of these conjugates by likely sequestering or extruding the hydrophobic or hydrophilic dyes, respectively. Application of the compounds for imaging collagen-rich tissue in an animal model of inflammatory arthritis showed enhanced uptake of all four conjugates, which retained high collagen-binding affinity, in inflamed joints. Moreover, LS1001 and LS1107 improved the arthritic joint-to-background contrast, suggesting that reduced aggregation enhanced the clearance of these compounds from non-target tissues. Our results highlight a peptide-driven strategy to alter the aggregation states of molecular probes in aqueous solutions, irrespective of the water-solubilizing properties of the dye molecules. The interplay between the monomeric and aggregated forms of the conjugates using simple thiol-modifiers lends the peptide-driven approach to diverse applications, including the effective imaging of inflammatory arthritis joints.
Collapse
Affiliation(s)
- Megan S. Michie
- Optical Radiology Laboratory, Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Baogang Xu
- Optical Radiology Laboratory, Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Gail Sudlow
- Optical Radiology Laboratory, Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Luke E. Springer
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Christine T.N. Pham
- Department of Medicine, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Samuel Achilefu
- Optical Radiology Laboratory, Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
25
|
Non-clinical safety assessment and in vivo biodistribution of CoviFab, an RBD-specific F(ab')2 fragment derived from equine polyclonal antibodies. Toxicol Appl Pharmacol 2022; 434:115796. [PMID: 34785274 PMCID: PMC8590615 DOI: 10.1016/j.taap.2021.115796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/16/2021] [Accepted: 11/10/2021] [Indexed: 12/23/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has required the urgent development of new therapies, among which passive immunotherapy is contemplated. CoviFab (INM005) is a RBD-specific F(ab′)2 fragment derived from equine polyclonal antibodies. We investigate their preclinical security and biodistribution by in vivo and ex vivo NIR imaging after intravenous administration of a dose of 4 mg/kg at time 0 and 48 h. Images were taken at 1, 12, 24, 36, 48, 49, 60, 72, 84, 96, 108, 120, 132 and 144 h after the first intravenous injection. At 96 and 144 h, mice were sacrificed for haematology, serum chemistry, clinical pathology, histopathology and ex vivo imaging. The biodistribution profile was similar in all organs studied, with the highest fluorescence at 1 h after each injection, gradually decreasing after that each one and until the end of the study (144 h). The toxicology study revealed no significant changes in the haematology and serum chemistry parameters. Further, there were no changes in the gross and histological examination of organs. Nonclinical data of the current study confirm that CoviFab is safe, without observable adverse effects in mice. Furthermore, we confirm that bioimaging studies are a useful approach in preclinical trials to determine biodistribution.
Collapse
|
26
|
van Leeuwen FW, van Willigen DM, Buckle T. Clinical application of fluorescent probes. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00104-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
27
|
Buckle T, van Willigen DM, Welling MM, van Leeuwen FW. Pre-clinical development of fluorescent tracers and translation towards clinical application. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00045-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
28
|
Vries HMD, Bekers E, van Oosterom MN, Karakullukcu MB, van HG, Poel D, van Leeuwen FWB, Buckle T, Brouwer OR. c-MET Receptor-Targeted Fluorescence on the Road to Image-Guided Surgery in Penile Squamous Cell Carcinoma Patients. J Nucl Med 2022; 63:51-56. [PMID: 33990404 PMCID: PMC8717176 DOI: 10.2967/jnumed.120.261864] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/20/2021] [Indexed: 11/16/2022] Open
Abstract
In penile squamous cell carcinoma (pSCC), primary surgery aims to obtain oncologically safe margins while minimizing mutilation. Surgical guidance provided by receptor-specific tracers could potentially improve margin detection and reduce unnecessary excision of healthy tissue. Here, we present the first results of a prospective feasibility study for real-time intraoperative visualization of pSCC using a fluorescent mesenchymal-epithelial transition factor (c-MET) receptor targeting tracer (EMI-137). Methods: EMI-137 tracer performance was initially assessed ex vivo (n = 10) via incubation of freshly excised pSCC in a solution containing EMI-137 (500 nM). The in vivo potential of c-MET targeting and intraoperative tumor visualization was assessed after intravenous administration of EMI-137 to 5 pSCC patients scheduled for surgical resection using a cyanine-5 fluorescence camera. Fluorescence imaging results were related to standard pathologic tumor evaluation and c-MET immunohistochemistry. Three of the 5 in vivo patients also underwent a sentinel node resection after local administration of the hybrid tracer indocyanine green- 99mTc-nanocolloid, which could be imaged using a near-infrared fluorescence camera. Results: No tracer-related adverse events were encountered. Both ex vivo and in vivo, EMI-137 enabled c-MET-based tumor visualization in all patients. Histopathologic analyses showed that all pSCCs expressed c-MET, with expression levels of at least 70% in 14 of 15 patients. Moreover, the highest c-MET expression levels were seen on the outside rim of the tumors, and a visual correlation was found between c-MET expression and fluorescence signal intensity. No complications were encountered when combining primary tumor targeting with lymphatic mapping. As such, simultaneous use of cyanine-5 and indocyanine green in the same patient proved to be feasible. Conclusion: Fluorescence imaging of c-MET receptor- expressing pSCC tumors after intravenous injection of EMI-137 was shown to be feasible and can be combined with fluorescence-based lymphatic mapping. This combination is unique and paves the way toward further development of this surgical guidance approach.
Collapse
Affiliation(s)
- Hielke M de Vries
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Elise Bekers
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands; and
| | - Matthias N van Oosterom
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - M Baris Karakullukcu
- Department of Head and Neck Surgery, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - der Poel
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Fijs W B van Leeuwen
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Tessa Buckle
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands;
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Oscar R Brouwer
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands;
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
29
|
Schouw HM, Huisman LA, Janssen YF, Slart RHJA, Borra RJH, Willemsen ATM, Brouwers AH, van Dijl JM, Dierckx RA, van Dam GM, Szymanski W, Boersma HH, Kruijff S. Targeted optical fluorescence imaging: a meta-narrative review and future perspectives. Eur J Nucl Med Mol Imaging 2021; 48:4272-4292. [PMID: 34633509 PMCID: PMC8566445 DOI: 10.1007/s00259-021-05504-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/23/2021] [Indexed: 12/27/2022]
Abstract
Purpose The aim of this review is to give an overview of the current status of targeted optical fluorescence imaging in the field of oncology, cardiovascular, infectious and inflammatory diseases to further promote clinical translation. Methods A meta-narrative approach was taken to systematically describe the relevant literature. Consecutively, each field was assigned a developmental stage regarding the clinical implementation of optical fluorescence imaging. Results Optical fluorescence imaging is leaning towards clinical implementation in gastrointestinal and head and neck cancers, closely followed by pulmonary, neuro, breast and gynaecological oncology. In cardiovascular and infectious disease, optical imaging is in a less advanced/proof of concept stage. Conclusion Targeted optical fluorescence imaging is rapidly evolving and expanding into the clinic, especially in the field of oncology. However, the imaging modality still has to overcome some major challenges before it can be part of the standard of care in the clinic, such as the provision of pivotal trial data. Intensive multidisciplinary (pre-)clinical joined forces are essential to overcome the delivery of such compelling phase III registration trial data and subsequent regulatory approval and reimbursement hurdles to advance clinical implementation of targeted optical fluorescence imaging as part of standard practice. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05504-y.
Collapse
Affiliation(s)
- H M Schouw
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - L A Huisman
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Y F Janssen
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - R H J A Slart
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - R J H Borra
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Radiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - A T M Willemsen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - A H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - J M van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - R A Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Diagnostic Sciences, Ghent University Faculty of Medicine and Health Sciences, Gent, Belgium
| | - G M van Dam
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,AxelaRx/TRACER Europe BV, Groningen, The Netherlands
| | - W Szymanski
- Department of Radiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - H H Boersma
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Centre of Groningen, Groningen, The Netherlands
| | - S Kruijff
- Department of Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands. .,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.
| |
Collapse
|
30
|
Şen Karaman D, Pamukçu A, Karakaplan MB, Kocaoglu O, Rosenholm JM. Recent Advances in the Use of Mesoporous Silica Nanoparticles for the Diagnosis of Bacterial Infections. Int J Nanomedicine 2021; 16:6575-6591. [PMID: 34602819 PMCID: PMC8478671 DOI: 10.2147/ijn.s273062] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Public awareness of infectious diseases has increased in recent months, not only due to the current COVID-19 outbreak but also because of antimicrobial resistance (AMR) being declared a top-10 global health threat by the World Health Organization (WHO) in 2019. These global issues have spiked the realization that new and more efficient methods and approaches are urgently required to efficiently combat and overcome the failures in the diagnosis and therapy of infectious disease. This holds true not only for current diseases, but we should also have enough readiness to fight the unforeseen diseases so as to avoid future pandemics. A paradigm shift is needed, not only in infection treatment, but also diagnostic practices, to overcome the potential failures associated with early diagnosis stages, leading to unnecessary and inefficient treatments, while simultaneously promoting AMR. With the development of nanotechnology, nanomaterials fabricated as multifunctional nano-platforms for antibacterial therapeutics, diagnostics, or both (known as "theranostics") have attracted increasing attention. In the research field of nanomedicine, mesoporous silica nanoparticles (MSN) with a tailored structure, large surface area, high loading capacity, abundant chemical versatility, and acceptable biocompatibility, have shown great potential to integrate the desired functions for diagnosis of bacterial infections. The focus of this review is to present the advances in mesoporous materials in the form of nanoparticles (NPs) or composites that can easily and flexibly accommodate dual or multifunctional capabilities of separation, identification and tracking performed during the diagnosis of infectious diseases together with the inspiring NP designs in diagnosis of bacterial infections.
Collapse
Affiliation(s)
- Didem Şen Karaman
- Biomedical Engineering Department, Faculty of Engineering and Architecture, İzmir Katip Çelebi University, İzmir, 35620, Turkey
| | - Ayşenur Pamukçu
- İzmir Kâtip Çelebi University, Graduate School of Natural and Applied Sciences, Department of Biomedical Technologies, İzmir, Turkey
| | - M Baran Karakaplan
- İzmir Kâtip Çelebi University, Graduate School of Natural and Applied Sciences, Department of Biomedical Engineering, İzmir, Turkey
| | - Ozden Kocaoglu
- Biomedical Engineering Department, Faculty of Engineering and Architecture, İzmir Katip Çelebi University, İzmir, 35620, Turkey
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
| |
Collapse
|
31
|
Han Z, Ke M, Liu X, Wang J, Guan Z, Qiao L, Wu Z, Sun Y, Sun X. Molecular Imaging, How Close to Clinical Precision Medicine in Lung, Brain, Prostate and Breast Cancers. Mol Imaging Biol 2021; 24:8-22. [PMID: 34269972 DOI: 10.1007/s11307-021-01631-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 12/15/2022]
Abstract
Precision medicine is playing a pivotal role in strategies of cancer therapy. Unlike conventional one-size-fits-all chemotherapy or radiotherapy modalities, precision medicine could customize an individual treatment plan for cancer patients to acquire superior efficacy, while minimizing side effects. Precision medicine in cancer therapy relies on precise and timely tumor biological information. Traditional tissue biopsies, however, are often inadequate in meeting this requirement due to cancer heterogeneity, poor tolerance, and invasiveness. Molecular imaging could detect tumor biology characterization in a noninvasive and visual manner, and provide information about therapeutic targets, treatment response, and pharmacodynamic evaluation. This summates to significant value in guiding cancer precision medicine in aspects of patient screening, treatment monitoring, and estimating prognoses. Although growing clinical evidences support the further application of molecular imaging in precision medicine of cancer, some challenges remain. In this review, we briefly summarize and discuss representative clinical trials of molecular imaging in improving precision medicine of cancer patients, aiming to provide useful references for facilitating further clinical translation of molecular imaging to precision medicine of cancers.
Collapse
Affiliation(s)
- Zhaoguo Han
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
- Biomedical Research Imaging Center, Department of Radiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Mingxing Ke
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xiang Liu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jing Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Zhengqi Guan
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Lina Qiao
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Zhexi Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Yingying Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xilin Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China.
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
32
|
Ghanim M, Relitti N, McManus G, Butini S, Cappelli A, Campiani G, Mok KH, Kelly VP. A non-toxic, reversibly released imaging probe for oral cancer that is derived from natural compounds. Sci Rep 2021; 11:14069. [PMID: 34234213 PMCID: PMC8263592 DOI: 10.1038/s41598-021-93408-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 06/24/2021] [Indexed: 02/06/2023] Open
Abstract
CD44 is emerging as an important receptor biomarker for various cancers. Amongst these is oral cancer, where surgical resection remains an essential mode of treatment. Unfortunately, surgery is frequently associated with permanent disfigurement, malnutrition, and functional comorbidities due to the difficultly of tumour removal. Optical imaging agents that can guide tumour tissue identification represent an attractive approach to minimising the impact of surgery. Here, we report the synthesis of a water-soluble fluorescent probe, namely HA-FA-HEG-OE (compound 1), that comprises components originating from natural sources: oleic acid, ferulic acid and hyaluronic acid. Compound 1 was found to be non-toxic, displayed aggregation induced emission and accumulated intracellularly in vesicles in SCC-9 oral squamous cells. The uptake of 1 was fully reversible over time. Internalization of compound 1 occurs through receptor mediated endocytosis; uniquely mediated through the CD44 receptor. Uptake is related to tumorigenic potential, with non-tumorigenic, dysplastic DOK cells and poorly tumorigenic MCF-7 cells showing only low intracellular levels and highlighting the critical role of endocytosis in cancer progression and metastasis. Together, the recognised importance of CD44 as a cancer stem cell marker in oral cancer, and the reversible, non-toxic nature of 1, makes it a promising agent for real time intraoperative imaging.
Collapse
Affiliation(s)
- Magda Ghanim
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Nicola Relitti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, 53100, Siena, Italy
| | - Gavin McManus
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, 53100, Siena, Italy.
| | - Andrea Cappelli
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, 53100, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, 53100, Siena, Italy
| | - K H Mok
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Vincent P Kelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
33
|
Wartak A, Kelada AK, Leon Alarcon PA, Bablouzian AL, Ahsen OO, Gregg AL, Wei Y, Bollavaram K, Sheil CJ, Farewell E, VanTol S, Smith R, Grahmann P, Baillargeon AR, Gardecki JA, Tearney GJ. Dual-modality optical coherence tomography and fluorescence tethered capsule endomicroscopy. BIOMEDICAL OPTICS EXPRESS 2021; 12:4308-4323. [PMID: 34457416 PMCID: PMC8367220 DOI: 10.1364/boe.422453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 06/13/2023]
Abstract
OCT tethered capsule endomicroscopy (TCE) is an emerging noninvasive diagnostic imaging technology for gastrointestinal (GI) tract disorders. OCT measures tissue reflectivity that provides morphologic image contrast, and thus is incapable of ascertaining molecular information that can be useful for improving diagnostic accuracy. Here, we introduce an extension to OCT TCE that includes a fluorescence (FL) imaging channel for attaining complementary, co-registered molecular contrast. We present the development of an OCT-FL TCE capsule and a portable, plug-and-play OCT-FL imaging system. The technology is validated in phantom experiments and feasibility is demonstrated in a methylene blue (MB)-stained swine esophageal injury model, ex vivo and in vivo.
Collapse
Affiliation(s)
- Andreas Wartak
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Alfred K. Kelada
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Paola A. Leon Alarcon
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ara L. Bablouzian
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Osman O. Ahsen
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Abigail L. Gregg
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Yuxiao Wei
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Keval Bollavaram
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Conor J. Sheil
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Edward Farewell
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Schuyler VanTol
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rachel Smith
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Patricia Grahmann
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Aaron R. Baillargeon
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joseph A. Gardecki
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Guillermo J. Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
34
|
Kang X, Li M, Liu L, Liu S, Hu H, Zhang R, Ning S, Tian Z, Pan Y, Guo X, Wu K. Targeted imaging of esophageal adenocarcinoma with a near-infrared fluorescent peptide. BMC Gastroenterol 2021; 21:260. [PMID: 34118882 PMCID: PMC8199829 DOI: 10.1186/s12876-021-01840-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 06/08/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Targeted optical imaging offers a noninvasive and accurate method for the early detection of gastrointestinal tumors, especially for flat appearances. In our previous study, a sequence of SNFYMPL (SNF) was identified as a specific peptide to bind to esophageal carcinoma using phage-display technology. This study aimed to evaluate the tumor-targeting efficacy of Cy5.5-conjugated SNF probe for imaging of esophageal carcinoma in vitro and in vivo. METHODS The SNF-Cy5.5 probe was synthesized and then identified using High Performance Liquid Chromatography (HPLC) and mass spectrometry (MS). Confocal fluorescence imaging and Flow cytometry analysis were performed to evaluate the binding specificity and the receptor binding affinity of SNF-Cy5.5 to OE33. In vivo imaging was performed to evaluate the targeting ability of SNF-Cy5.5 to esophageal carcinoma. RESULTS The confocal imaging and flow cytometry analysis showed that SNF-Cy5.5 bound specifically to the plasma membrane of OE33 cells with a high affinity. In vivo, for non-block group, SNF-Cy5.5 probe exhibited rapid OE33 tumor targeting during 24 h p.i. and excellent tumor-to-background contrast at 2 h p.i. For the block group, SNF-Cy5.5 was not observed in the mice after 4 h p.i. Ex vivo imaging also revealed that a higher fluorescent signal intensity value of the tumors was clearly observed in the non-block group than that in the block group (2.6 ± 0.32 × 109 vs. 0.8 ± 0.08 × 109, p < 0.05). CONCLUSIONS SNF-Cy5.5 was synthesized and characterized with a high efficiency and purity. The higher affinity, specificity, and tumor targeting efficacy of SNF-Cy5.5 were confirmed by in vitro and in vivo tests. SNF-Cy5.5 is a promising optical probe for the imaging of esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Xiaoyu Kang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Meng Li
- Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Lei Liu
- Department of Gastroenterology, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shaopeng Liu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Hao Hu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Rui Zhang
- Department of Critical Care Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi, People's Republic of China.,College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Siming Ning
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Zuhong Tian
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Yanglin Pan
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, People's Republic of China.
| | - Xuegang Guo
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, People's Republic of China
| |
Collapse
|
35
|
Buckle T, van Alphen M, van Oosterom MN, van Beurden F, Heimburger N, van der Wal JE, van den Brekel M, van Leeuwen FWB, Karakullukcu B. Translation of c-Met Targeted Image-Guided Surgery Solutions in Oral Cavity Cancer-Initial Proof of Concept Data. Cancers (Basel) 2021; 13:cancers13112674. [PMID: 34071623 PMCID: PMC8198422 DOI: 10.3390/cancers13112674] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Translation of tumor-specific fluorescent tracers is crucial in the realization intraoperative of tumor identification during fluorescence-guided surgery. Ex vivo assessment of surgical specimens after topical tracer application has the potential to reveal the suitability of a potential surgical target prior to in vivo use in patients. In this study, the c-Met receptor was identified as a possible candidate for fluorescence-guided surgery in oral cavity cancer. Freshly excised tumor specimens obtained from ten patients with squamous cell carcinoma of the tongue were incubated with EMI-137 and imaged with a clinical-grade Cy5 prototype fluorescence camera. In total, 9/10 tumors were fluorescently illuminated, while non-visualization could be linked to non-superficial tumor localization. Immunohistochemistry revealed c-Met expression in all ten specimens. Tumor assessment was improved via video representation of the tumor-to-background ratio. Abstract Intraoperative tumor identification (extension/margins/metastases) via receptor-specific targeting is one of the ultimate promises of fluorescence-guided surgery. The translation of fluorescent tracers that enable tumor visualization forms a critical component in the realization of this approach. Ex vivo assessment of surgical specimens after topical tracer application could help provide an intermediate step between preclinical evaluation and first-in-human trials. Here, the suitability of the c-Met receptor as a potential surgical target in oral cavity cancer was explored via topical ex vivo application of the fluorescent tracer EMI-137. Freshly excised tumor specimens obtained from ten patients with squamous cell carcinoma of the tongue were incubated with EMI-137 and imaged with a clinical-grade Cy5 prototype fluorescence camera. In-house developed image processing software allowed video-rate assessment of the tumor-to-background ratio (TBR). Fluorescence imaging results were related to standard pathological evaluation and c-MET immunohistochemistry. After incubation with EMI-137, 9/10 tumors were fluorescently illuminated. Immunohistochemistry revealed c-Met expression in all ten specimens. Non-visualization could be linked to a more deeply situated lesion. Tumor assessment was improved via video representation of the TBR (median TBR: 2.5 (range 1.8–3.1)). Ex vivo evaluation of tumor specimens suggests that c-Met is a possible candidate for fluorescence-guided surgery in oral cavity cancer.
Collapse
Affiliation(s)
- Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.N.v.O.); (F.v.B.); (N.H.); (F.W.B.v.L.)
- Department of Head and Neck Surgery and Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands; (M.v.A.); (M.v.d.B.); (B.K.)
- Correspondence:
| | - Maarten van Alphen
- Department of Head and Neck Surgery and Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands; (M.v.A.); (M.v.d.B.); (B.K.)
| | - Matthias N. van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.N.v.O.); (F.v.B.); (N.H.); (F.W.B.v.L.)
- Department of Head and Neck Surgery and Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands; (M.v.A.); (M.v.d.B.); (B.K.)
| | - Florian van Beurden
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.N.v.O.); (F.v.B.); (N.H.); (F.W.B.v.L.)
| | - Nina Heimburger
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.N.v.O.); (F.v.B.); (N.H.); (F.W.B.v.L.)
| | - Jaqueline E. van der Wal
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands;
| | - Michiel van den Brekel
- Department of Head and Neck Surgery and Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands; (M.v.A.); (M.v.d.B.); (B.K.)
| | - Fijs W. B. van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (M.N.v.O.); (F.v.B.); (N.H.); (F.W.B.v.L.)
- Department of Head and Neck Surgery and Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands; (M.v.A.); (M.v.d.B.); (B.K.)
| | - Baris Karakullukcu
- Department of Head and Neck Surgery and Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, The Netherlands; (M.v.A.); (M.v.d.B.); (B.K.)
| |
Collapse
|
36
|
Böhmer VI, Szymanski W, Feringa BL, Elsinga PH. Multivalent Probes in Molecular Imaging: Reality or Future? Trends Mol Med 2021; 27:379-393. [PMID: 33436332 DOI: 10.1016/j.molmed.2020.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/17/2020] [Accepted: 12/08/2020] [Indexed: 01/25/2023]
Abstract
The rapidly developing field of molecular medical imaging focuses on specific visualization of (patho)physiological processes through the application of imaging agents (IAs) in multiple clinical modalities. Although our understanding of the principles underlying efficient IAs design has increased tremendously, many IAs still show poor in vivo imaging performance because of low binding affinity and/or specificity. These limitations can be addressed by taking advantage of multivalency, in which multiple copies of a ligand are employed to strengthen the interaction. We critically address specific challenges associated with the application of multivalent compounds in molecular imaging, and we give directions for a stepwise approach to the design of multivalent imaging probes to improve their target binding and pharmacokinetics (PK) for improved diagnostic potential.
Collapse
Affiliation(s)
- Verena I Böhmer
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands; Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747, AF, Groningen, The Netherlands
| | - Wiktor Szymanski
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747, AF, Groningen, The Netherlands; Department of Radiology, Medical Imaging Center, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands
| | - Ben L Feringa
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, 9747, AF, Groningen, The Netherlands
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands.
| |
Collapse
|
37
|
Lauwerends LJ, van Driel PBAA, Baatenburg de Jong RJ, Hardillo JAU, Koljenovic S, Puppels G, Mezzanotte L, Löwik CWGM, Rosenthal EL, Vahrmeijer AL, Keereweer S. Real-time fluorescence imaging in intraoperative decision making for cancer surgery. Lancet Oncol 2021; 22:e186-e195. [PMID: 33765422 DOI: 10.1016/s1470-2045(20)30600-8] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023]
Abstract
Fluorescence-guided surgery is an intraoperative optical imaging method that provides surgeons with real-time guidance for the delineation of tumours. Currently, in phase 1 and 2 clinical trials, evaluation of fluorescence-guided surgery is primarily focused on its diagnostic performance, although the corresponding outcome variables do not inform about the added clinical benefit of fluorescence-guided surgery and are challenging to assess objectively. Nonetheless, the effect of fluorescence-guided surgery on intraoperative decision making is the most objective outcome measurement to assess the clinical value of this imaging method. In this Review, we explore the study designs of existing trials of fluorescence-guided surgery that allow us to extract information on potential changes in intraoperative decision making, such as additional or more conservative resections. On the basis of this analysis, we offer recommendations on how to report changes in intraoperative decision making that result from fluorescence imaging, which is of utmost importance for the widespread clinical implementation of fluorescence-guided surgery.
Collapse
Affiliation(s)
- Lorraine J Lauwerends
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus University Medical Center, Rotterdam, Netherlands; Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Robert J Baatenburg de Jong
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus University Medical Center, Rotterdam, Netherlands; Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - José A U Hardillo
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus University Medical Center, Rotterdam, Netherlands; Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Senada Koljenovic
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands; Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Gerwin Puppels
- Department of Dermatology, Erasmus University Medical Center, Rotterdam, Netherlands; Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, Netherlands; Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Clemens W G M Löwik
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, Netherlands; Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Stijn Keereweer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus University Medical Center, Rotterdam, Netherlands; Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
38
|
Not so innocent: Impact of fluorophore chemistry on the in vivo properties of bioconjugates. Curr Opin Chem Biol 2021; 63:38-45. [PMID: 33684856 DOI: 10.1016/j.cbpa.2021.01.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 12/11/2022]
Abstract
The combination of targeting ligands and fluorescent dyes is a powerful strategy to observe cell types and tissues of interest. Conjugates of peptides, proteins, and, in particular, monoclonal antibodies (mAbs) exhibit excellent tumor targeting in various contexts. This approach has been translated to a clinical setting to provide real-time molecular insights during the surgical resection of solid tumors. A critical element of this approach is the generation of highly fluorescent bioconjugates that maintain the properties of the parent targeting ligand. A number of studies have found that fluorophores can dramatically impact the pharmacokinetic and tumor-targeting properties of the bioconjugates they are meant to only innocently observe. In this review, we summarize several examples of these effects and highlight strategies that have been used to mitigate them. These include the application of site-specific labeling chemistries, modulating label density, and altering the structure of the fluorescent probe itself. In particular, we point out the significant potential of fluorophores with hydrophilic but net-neutral structures. Overall, this review highlights recent progress in refining the in vivo properties of fluorescent bioconjugates, and we hope, will inform future efforts in this area.
Collapse
|
39
|
Lee YJ, Krishnan G, Nishio N, van den Berg NS, Lu G, Martin BA, van Keulen S, Colevas AD, Kapoor S, Liu JTC, Rosenthal EL. Intraoperative Fluorescence-Guided Surgery in Head and Neck Squamous Cell Carcinoma. Laryngoscope 2021; 131:529-534. [PMID: 33593036 DOI: 10.1002/lary.28822] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023]
Abstract
The rate of positive margins in head and neck cancers has remained stagnant over the past three decades and is consistently associated with poor overall survival. This suggests that significant improvements must be made intraoperatively to ensure negative margins. We discuss the important role of fluorescence imaging to guide surgical oncology in head and neck cancer. This review includes a general overview of the principles of fluorescence, available fluorophores used for fluorescence imaging, and specific clinical applications of fluorescence-guided surgery, as well as challenges and future directions in head and neck surgical oncology. Laryngoscope, 131:529-534, 2021.
Collapse
Affiliation(s)
- Yu-Jin Lee
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Giri Krishnan
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A.,Department of Otolaryngology, Head and Neck Surgery, University of Adelaide, Adelaide, SA, Australia
| | - Naoki Nishio
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Nynke S van den Berg
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Guolan Lu
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Brock A Martin
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Stan van Keulen
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Alexander D Colevas
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Shrey Kapoor
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A
| | - Jonathan T C Liu
- Department of Mechanical Engineering, University of Washington, Seattle, WA, U.S.A.,Department of Bioengineering, University of Washington, Seattle, WA, U.S.A.,Department of Pathology, University of Washington, Seattle, WA, U.S.A
| | - Eben L Rosenthal
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, U.S.A.,Department of Radiology, Stanford University School of Medicine, Stanford, CA, U.S.A
| |
Collapse
|
40
|
Salinas HR, Miyasato DL, Eremina OE, Perez R, Gonzalez KL, Czaja AT, Burkitt S, Aron A, Fernando A, Ojeda LS, Larson KN, Mohamed AW, Campbell JL, Goins BA, Zavaleta C. A colorful approach towards developing new nano-based imaging contrast agents for improved cancer detection. Biomater Sci 2021; 9:482-495. [PMID: 32812951 PMCID: PMC7855687 DOI: 10.1039/d0bm01099e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Providing physicians with new imaging agents to help detect cancer with better sensitivity and specificity has the potential to significantly improve patient outcomes. Development of new imaging agents could offer improved early cancer detection during routine screening or help surgeons identify tumor margins for surgical resection. In this study, we evaluate the optical properties of a colorful class of dyes and pigments that humans routinely encounter. The pigments are often used in tattoo inks and the dyes are FDA approved for the coloring of foods, drugs, and cosmetics. We characterized their absorption, fluorescence and Raman scattering properties in the hopes of identifying a new panel of dyes that offer exceptional imaging contrast. We found that some of these coloring agents, coined as "optical inks", exhibit a multitude of useful optical properties, outperforming some of the clinically approved imaging dyes on the market. The best performing optical inks (Green 8 and Orange 16) were further incorporated into liposomal nanoparticles to assess their tumor targeting and optical imaging potential. Mouse xenograft models of colorectal, cervical and lymphoma tumors were used to evaluate the newly developed nano-based imaging contrast agents. After intravenous injection, fluorescence imaging revealed significant localization of the new "optical ink" liposomal nanoparticles in all three tumor models as opposed to their neighboring healthy tissues (p < 0.05). If further developed, these coloring agents could play important roles in the clinical setting. A more sensitive imaging contrast agent could enable earlier cancer detection or help guide surgical resection of tumors, both of which have been shown to significantly improve patient survival.
Collapse
Affiliation(s)
- Helen R Salinas
- Department of Biomedical Engineering, University of Southern California, 1002 Childs Way, Los Angeles, CA 90089, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Alramdan MHA, Kasalak Ö, Been LB, Suurmeijer AJH, Yakar D, Kwee TC. MRI after Whoops procedure: diagnostic value for residual sarcoma and predictive value for an incomplete second resection. Skeletal Radiol 2021; 50:2213-2220. [PMID: 33900432 PMCID: PMC8449770 DOI: 10.1007/s00256-021-03790-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine the value of MRI for the detection and assessment of the anatomic extent of residual sarcoma after a Whoops procedure (unplanned sarcoma resection) and its utility for the prediction of an incomplete second resection. MATERIALS AND METHODS This study included consecutive patients who underwent a Whoops procedure, successively followed by gadolinium chelate-enhanced MRI and second surgery at a tertiary care sarcoma center. RESULTS Twenty-six patients were included, of whom 19 with residual tumor at the second surgery and 8 with an incomplete second resection (R1: n = 6 and R2: n = 2). Interobserver agreement for residual tumor at MRI after a Whoops procedure was perfect (κ value: 1.000). MRI achieved a sensitivity of 47.4% (9/19), a specificity of 100% (7/7), a positive predictive value of 100% (9/9), and a negative predictive value of 70.0% (7/17) for the detection of residual tumor. MRI correctly classified 2 of 19 residual sarcomas as deep-seated (i.e., extending beyond the superficial muscle fascia) but failed to correctly classify 3 of 19 residual sarcomas as deep-seated. There were no significant associations between MRI findings (presence of residual tumor, maximum tumor diameter, anatomic tumor extent, tumor margins, tumor spiculae, and tumor tail on the superficial fascia) with an incomplete (R1 or R2) second resection. CONCLUSION Gadolinium chelate-enhanced MRI is a reproducible method to rule in residual sarcoma, but it is insufficiently accurate to rule out and assess the anatomic extent or residual sarcoma after a Whoops procedure. Furthermore, MRI has no utility in predicting an incomplete second resection.
Collapse
Affiliation(s)
- Mohammed H. A. Alramdan
- grid.4494.d0000 0000 9558 4598Department of Radiology, Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Ömer Kasalak
- grid.4494.d0000 0000 9558 4598Department of Radiology, Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Lukas B. Been
- grid.4494.d0000 0000 9558 4598Department of Surgical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert J. H. Suurmeijer
- grid.4494.d0000 0000 9558 4598Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Derya Yakar
- grid.4494.d0000 0000 9558 4598Department of Radiology, Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | - Thomas C. Kwee
- grid.4494.d0000 0000 9558 4598Department of Radiology, Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
42
|
The Emerging Role of CD24 in Cancer Theranostics-A Novel Target for Fluorescence Image-Guided Surgery in Ovarian Cancer and Beyond. J Pers Med 2020; 10:jpm10040255. [PMID: 33260974 PMCID: PMC7712410 DOI: 10.3390/jpm10040255] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
Complete cytoreductive surgery is the cornerstone of the treatment of epithelial ovarian cancer (EOC). The application of fluorescence image-guided surgery (FIGS) allows for the increased intraoperative visualization and delineation of malignant lesions by using fluorescently labeled targeting biomarkers, thereby improving intraoperative guidance. CD24, a small glycophosphatidylinositol-anchored cell surface receptor, is overexpressed in approximately 70% of solid cancers, and has been proposed as a prognostic and therapeutic tumor-specific biomarker for EOC. Recently, preclinical studies have demonstrated the benefit of CD24-targeted contrast agents for non-invasive fluorescence imaging, as well as improved tumor resection by employing CD24-targeted FIGS in orthotopic patient-derived xenograft models of EOC. The successful detection of miniscule metastases denotes CD24 as a promising biomarker for the application of fluorescence-guided surgery in EOC patients. The aim of this review is to present the clinical and preclinically evaluated biomarkers for ovarian cancer FIGS, highlight the strengths of CD24, and propose a future bimodal approach combining CD24-targeted fluorescence imaging with radionuclide detection and targeted therapy.
Collapse
|
43
|
Zhang D, Huang J, Li W, Zhang Z, Zhu M, Feng Y, Zhao Y, Li Y, Lu S, He S. Screening and identification of a CD44v6 specific peptide using improved phage display for gastric cancer targeting. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1442. [PMID: 33313187 PMCID: PMC7723568 DOI: 10.21037/atm-19-4781] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Peptide probes can be applied for biomarker targeting to improve the diagnostic accuracy. Cluster of differentiation 44 (CD44) is up-regulated in gastric cancer (GC). Among all the variants of CD44, CD44v6 is reported the most promising biomarker for GC. The purpose of this study was generating and identification a peptide ligand specific to CD44v6. Methods A 12-mer phage peptide library was screened on CD44v overexpressed HEK-293 cells with an improved subtractive method. Five candidate sequences emerged. Candidate phages were selected using enzyme-linked immunosorbent assay and competitive inhibition assays. Then the sequence (designated ELT) was chosen for further study. Its binding affinity and specificity were verified on recombinant protein, GC cells, GC tissues and xenograft models based on BALB/c-nu/nu mice using dissociation constant calculation, immunofluorescence, immunohistochemistry and in vivo imaging separately. Results The dissociation constant of ELT with recombinant protein was 611.2 nM. ELT stained CD44v overexpressed HEK-293 but not the cell expressing wild-type CD44s. On GC cell lines, ELT co-stained with anti-CD44v6 antibody. ELT binding on tumor tissues significantly increased compared with that of paracancer tissues, also showed a linear positive correlation with CD44v6 expression. ELT specifically accumulated in tumor and eliminated in short time in vivo. Conclusions ELT can target GC in vitro and in vivo via CD44v6, indicating its potential to serve as a probe for GC targeting diagnosis and therapy.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Huang
- School of Electronic Information and Artificial Intelligence, Shannxi University of Science &Technology, Xi'an, China.,School of Materials Science and Engineering, Shannxi Normal University, Xi'an, China
| | - Weiming Li
- Department of Vascular Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhiyong Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Zhu
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yun Feng
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhao
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yarui Li
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shaoying Lu
- Department of Vascular Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuixiang He
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
44
|
Image-guided in situ detection of bacterial biofilms in a human prosthetic knee infection model: a feasibility study for clinical diagnosis of prosthetic joint infections. Eur J Nucl Med Mol Imaging 2020; 48:757-767. [PMID: 32901352 PMCID: PMC8036220 DOI: 10.1007/s00259-020-04982-w] [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: 04/18/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Due to an increased human life expectancy, the need to replace arthritic or dysfunctional joints by prosthetics is higher than ever before. Prosthetic joints are unfortunately inherently susceptible to bacterial infection accompanied by biofilm formation. Accurate and rapid diagnosis is vital to increase therapeutic success. Yet, established diagnostic modalities cannot directly detect bacterial biofilms on prostheses. Therefore, the present study was aimed at investigating whether arthroscopic optical imaging can accurately detect bacterial biofilms on prosthetic joints. METHODS Here, we applied a conjugate of the antibiotic vancomycin and the near-infrared fluorophore IRDye800CW, in short vanco-800CW, in combination with arthroscopic optical imaging to target and visualize biofilms on infected prostheses. RESULTS We show in a human post-mortem prosthetic knee infection model that a staphylococcal biofilm is accurately detected in real time and distinguished from sterile sections in high resolution. In addition, we demonstrate that biofilms associated with the clinically most relevant bacterial species can be detected using vanco-800CW. CONCLUSION The presented image-guided arthroscopic approach provides direct visual diagnostic information and facilitates immediate appropriate treatment selection.
Collapse
|
45
|
Cekanova M, Pandey S, Olin S, Ryan P, Stokes JE, Hecht S, Martin-Jimenez T, Uddin MJ, Marnett LJ. Pharmacokinetic characterization of fluorocoxib D, a cyclooxygenase-2-targeted optical imaging agent for detection of cancer. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200044R. [PMID: 32860356 PMCID: PMC7456637 DOI: 10.1117/1.jbo.25.8.086005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/11/2020] [Indexed: 06/11/2023]
Abstract
SIGNIFICANCE Fluorocoxib D, N-[(rhodamin-X-yl)but-4-yl]-2-[1-(4-chlorobenzoyl)-5-methoxy-2-methyl-1H-indol-3-yl]acetamide, is a water-soluble optical imaging agent to detect cyclooxygenase-2 (COX-2)-expressing cancer cells. AIM We evaluated the pharmacokinetic and safety properties of fluorocoxib D and its ability to detect cancer cells in vitro and in vivo. APPROACH Pharmacokinetic parameters of fluorocoxib D were assessed from plasma collected at designated time points after intravenous administration of 1 mg / kg fluorocoxib D in six research dogs using a high-performance liquid chromatography analysis. Safety of fluorocoxib D was assessed for 3 days after its administration using physical assessment, complete blood count, serum chemistry profile, and complete urinalysis in six research dogs. The ability of fluorocoxib D to detect COX-2-expressing cancer cells was performed using human 5637 cells in vitro and during rhinoscopy evaluation of specific fluorocoxib D uptake by canine cancer cells in vivo. RESULTS No evidence of toxicity and no clinically relevant adverse events were noted in dogs. Peak concentration of fluorocoxib D (114.8 ± 50.5 ng / ml) was detected in plasma collected at 0.5 h after its administration. Pretreatment of celecoxib blocked specific uptake of fluorocoxib D in COX-2-expressing human 5637 cancer cells. Fluorocoxib D uptake was detected in histology-confirmed COX-2-expressing head and neck cancer during rhinoscopy in a client-owned dog in vivo. Specific tumor-to-normal tissue ratio of detected fluorocoxib D signal was in an average of 3.7 ± 0.9 using Image J analysis. CONCLUSIONS Our results suggest that fluorocoxib D is a safe optical imaging agent used for detection of COX-2-expressing cancers and their margins during image-guided minimally invasive biopsy and surgical procedures.
Collapse
Affiliation(s)
- Maria Cekanova
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
- The University of Tennessee, UT-ORNL Graduate School of Genome, Science and Technology, Knoxville, Tennessee, United States
| | - Sony Pandey
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Shelly Olin
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Phillip Ryan
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Jennifer E. Stokes
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Silke Hecht
- The University of Tennessee, College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Knoxville, Tennessee, United States
| | - Tomas Martin-Jimenez
- The University of Tennessee, College of Veterinary Medicine, Department of Biomedical and Diagnostic Sciences, Knoxville, Tennessee, United States
| | - Md. Jashim Uddin
- Vanderbilt University School of Medicine, Vanderbilt Institute of Chemical Biology, Center for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry and Pharmacology, Nashville, Tennessee, United States
| | - Lawrence J. Marnett
- Vanderbilt University School of Medicine, Vanderbilt Institute of Chemical Biology, Center for Molecular Toxicology and Vanderbilt-Ingram Cancer Center, A. B. Hancock, Jr., Memorial Laboratory for Cancer Research, Departments of Biochemistry, Chemistry and Pharmacology, Nashville, Tennessee, United States
| |
Collapse
|
46
|
Evaluation of EphB4 as Target for Image-Guided Surgery of Breast Cancer. Pharmaceuticals (Basel) 2020; 13:ph13080172. [PMID: 32751634 PMCID: PMC7464973 DOI: 10.3390/ph13080172] [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: 06/18/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/02/2022] Open
Abstract
Background: Targeted image-guided surgery is based on the detection of tumor cells after administration of a radio-active or fluorescent tracer. Hence, enhanced binding of a tracer to tumor tissue compared to healthy tissue is crucial. Various tumor antigens have been evaluated as possible targets for image-guided surgery of breast cancer, with mixed results. Methods: In this study we have evaluated tyrosine kinase receptor EphB4, a member from the Eph tyrosine kinase receptor family, as a possible target for image-guided surgery of breast cancers. Two independent tissue micro arrays, consisting of matched sets of tumor and normal breast tissue, were stained for EphB4 by immunohistochemistry. The intensity of staining and the percentage of stained cells were scored by two independent investigators. Results: Immunohistochemical staining for EphB4 shows that breast cancer cells display enhanced membranous expression compared to adjacent normal breast tissue. The enhanced tumor staining is not associated with clinical variables like age of the patient or stage or subtype of the tumor, including Her2-status. Conclusion: These data suggest that EphB4 is a promising candidate for targeted image-guided surgery of breast cancer, especially for Her2 negative cases.
Collapse
|
47
|
Manwar R, Kratkiewicz K, Avanaki K. Overview of Ultrasound Detection Technologies for Photoacoustic Imaging. MICROMACHINES 2020; 11:E692. [PMID: 32708869 PMCID: PMC7407969 DOI: 10.3390/mi11070692] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022]
Abstract
Ultrasound detection is one of the major components of photoacoustic imaging systems. Advancement in ultrasound transducer technology has a significant impact on the translation of photoacoustic imaging to the clinic. Here, we present an overview on various ultrasound transducer technologies including conventional piezoelectric and micromachined transducers, as well as optical ultrasound detection technology. We explain the core components of each technology, their working principle, and describe their manufacturing process. We then quantitatively compare their performance when they are used in the receive mode of a photoacoustic imaging system.
Collapse
Affiliation(s)
- Rayyan Manwar
- Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48201, USA;
| | - Karl Kratkiewicz
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48201, USA;
| | - Kamran Avanaki
- Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48201, USA;
- Department of Dermatology, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
48
|
A review of optical breast imaging: Multi-modality systems for breast cancer diagnosis. Eur J Radiol 2020; 129:109067. [PMID: 32497943 DOI: 10.1016/j.ejrad.2020.109067] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 11/24/2022]
Abstract
This review of optical breast imaging describes basic physical and system principles and summarizes technological evolution with a focus on multi-modality platforms and recent clinical trial results. Ultrasound-guided diffuse optical tomography and co-registered ultrasound and photoacoustic imaging systems are emphasized as models of state of the art optical technology that are most conducive to clinical translation.
Collapse
|
49
|
CD24-targeted fluorescence imaging in patient-derived xenograft models of high-grade serous ovarian carcinoma. EBioMedicine 2020; 56:102782. [PMID: 32454401 PMCID: PMC7248428 DOI: 10.1016/j.ebiom.2020.102782] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/02/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The survival rate of patients with advanced high-grade serous ovarian carcinoma (HGSOC) remains disappointing. Clinically translatable orthotopic cell line xenograft models and patient-derived xenografts (PDXs) may aid the implementation of more personalised treatment approaches. Although orthotopic PDX reflecting heterogeneous molecular subtypes are considered the most relevant preclinical models, their use in therapeutic development is limited by lack of appropriate imaging modalities. METHODS We developed novel orthotopic xenograft and PDX models for HGSOC, and applied a near-infrared fluorescently labelled monoclonal antibody targeting the cell surface antigen CD24 for non-invasive molecular imaging of epithelial ovarian cancer. CD24-Alexa Fluor 680 fluorescence imaging was compared to bioluminescence imaging in three orthotopic cell line xenograft models of ovarian cancer (OV-90luc+, Skov-3luc+ and Caov-3luc+, n = 3 per model). The application of fluorescence imaging to assess treatment efficacy was performed in carboplatin-paclitaxel treated orthotopic OV-90 xenografts (n = 10), before the probe was evaluated to detect disease progression in heterogenous PDX models (n = 7). FINDINGS Application of the near-infrared probe, CD24-AF680, enabled both spatio-temporal visualisation of tumour development, and longitudinal therapy monitoring of orthotopic xenografts. Notably, CD24-AF680 facilitated imaging of multiple PDX models representing different histological subtypes of the disease. INTERPRETATION The combined implementation of CD24-AF680 and orthotopic PDX models creates a state-of-the-art preclinical platform which will impact the identification and validation of new targeted therapies, fluorescence image-guided surgery, and ultimately the outcome for HGSOC patients. FUNDING This study was supported by the H2020 program MSCA-ITN [675743], Helse Vest RHF, and Helse Bergen HF [911809, 911852, 912171, 240222, HV1269], as well as by The Norwegian Cancer Society [182735], and The Research Council of Norway through its Centers of excellence funding scheme [223250, 262652].
Collapse
|
50
|
Kwak MH, Yi G, Yang SM, Choe Y, Choi S, Lee HS, Kim E, Lim YB, Na K, Choi MG, Koo H, Park JM. A Dodecapeptide Selected by Phage Display as a Potential Theranostic Probe for Colon Cancers. Transl Oncol 2020; 13:100798. [PMID: 32454443 PMCID: PMC7248426 DOI: 10.1016/j.tranon.2020.100798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/23/2022] Open
Abstract
Aim Colon cancer is one of the leading causes of cancer-related mortality. However, specific biomarkers for its diagnosis or treatment are not established well. Methods We developed a colon-cancer specific peptide probe using phage display libraries. We validated the specificity of this probe to colon cancer cells with immunohistochemical staining and FACS analysis using one normal cell and five colon cancer cell lines. Results This peptide probe maintained binding affinity even after serum incubation. For therapeutic applications, this peptide probe was conjugated to hematoporphyrin, a photosensitizer, which showed a significantly enhanced cellular uptake and high photodynamic effect to kill tumor cells. As another application, we made a nanoparticle modified from the peptide probe. It efficiently delivered SN-38, an anticancer drug, into tumor cells, and its tumor-targeting ability was observed in vivo after intravenous injection to the same xenograft model. Conclusion The noble dodecapeptide probe can be a promising candidate for both colon tumor diagnosis and targeted drug delivery.
Collapse
Affiliation(s)
- Moon Hwa Kwak
- Catholic Photomedicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Gawon Yi
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Mok Yang
- Catholic Photomedicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Younghee Choe
- Catholic Photomedicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sangkee Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Hye-Soo Lee
- Department of Materials Science and Engineering, Yonsei University, Seoul, Republic of Korea
| | - Eunha Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Yong-Beom Lim
- Department of Materials Science and Engineering, Yonsei University, Seoul, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, Seoul, Republic of Korea
| | - Myung-Gyu Choi
- Catholic Photomedicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea; Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Jae Myung Park
- Catholic Photomedicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea; Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|