1
|
Kuronuma Y, Watanabe R, Hiruta Y. The latest developments of near-infrared fluorescent probes from NIR-I to NIR-II for bioimaging. ANAL SCI 2025:10.1007/s44211-025-00735-7. [PMID: 40019707 DOI: 10.1007/s44211-025-00735-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/09/2025] [Indexed: 03/01/2025]
Abstract
Near-infrared I (NIR-I: 650-950 nm) fluorescence imaging is a powerful tool for deep-tissue biological imaging, addressing the limitations of photon penetration depth in the visible-light region. Over the past decade, NIR imaging has extended to the near-infrared II (NIR-II: 1000-1700 nm) region, offering high-resolution and low background imaging by suppressing light scattering, and autofluorescence of tissues. Near-infrared fluorescent probes from NIR-I to NIR-II, with diverse functionalities, are increasingly utilized across biological fields to meet various detection needs and to explore physiological events in real time and spatial dimensions. This review discusses recent advancements in small-molecule NIR fluorescent dyes and probes, particularly those based on cyanine and rhodamine scaffolds, highlighting examples of their applications in bioimaging.
Collapse
Affiliation(s)
- Yuzuka Kuronuma
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Ryuji Watanabe
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan
| | - Yuki Hiruta
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa, 223-8522, Japan.
| |
Collapse
|
2
|
Tang J, Liao L, Xiao B, Sui Q, Zheng M, Jiang W, Han K, Kong L, Pan Z, Ding P. Efficacy and safety of subtotal pelvic peritonectomy for colorectal cancer patients with peritoneal metastasis confined to the pelvic cavity. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109703. [PMID: 40022888 DOI: 10.1016/j.ejso.2025.109703] [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: 08/08/2024] [Revised: 12/21/2024] [Accepted: 02/12/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Cytoreductive surgery has shown survival benefits for colorectal cancer (CRC) patients with peritoneal metastasis. However, the optimal extent of peritonectomy remains controversial in cases of limited peritoneal metastases. This study modified selective pelvic peritonectomy (SPP) into subtotal pelvic peritonectomy (STPP) for metastasis confined to pelvic cavity, and aimed to evaluate its feasibility, safety, and impact on survival outcomes. MATERIALS AND METHODS CRC patients with limited peritoneal metastasis confined to the pelvic cavity who underwent CC0 (no macroscopic residual cancer remained) resection were included from a prospectively collected database. Surgical complications, disease-free survival (DFS), and overall survival (OS) were analyzed. RESULTS A total of 67 patients were included (26 in the STPP group and 41 in the SPP group). Clinically, STPP was found to be feasible and without increased surgical complications or mortality rates. At a median follow-up of 33.9 months, the 3-year DFS was 65.9 % and 30.7 % in STPP and SPP groups, respectively (P= 0.002). The 3-year OS was 84.1 % and 68.5 % in STPP and SPP groups, respectively (P= 0.006). Moreover, STTP was independently associated with improved DFS (HR = 0.351, 95 % CI 0.165-0.745, P= 0.006) and OS (HR = 0.324, 95 % CI 0.116-0.902, P=0.032). Female gender was also independently associated with poor DFS (HR = 2.146, 95 % CI 1.078-4.271, P= 0.031). Among 24 female patients with remaining ovaries, 9 (37.5 %) cases developed metachronous ovarian metastasis, and of these 6 underwent a second operation. CONCLUSIONS Subtotal pelvic peritonectomy is associated with promising long-term outcomes in CRC patients with peritoneal metastasis confined to the pelvic cavity. Prophylactic bilateral oophorectomy should be strongly considered during cytoreductive surgery.
Collapse
Affiliation(s)
- Jinghua Tang
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China
| | - Leen Liao
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China
| | - Binyi Xiao
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China
| | - Qiaoqi Sui
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China
| | - Muxu Zheng
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China
| | - Wu Jiang
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China
| | - Kai Han
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China
| | - Lingheng Kong
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China
| | - Zhizhong Pan
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China.
| | - Peirong Ding
- Departments of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, 510060, PR China.
| |
Collapse
|
3
|
Martinelli S, Fortuna L, Coratti F, Passagnoli F, Amedei A, Cianchi F. Potential Probes for Targeted Intraoperative Fluorescence Imaging in Gastric Cancer. Cancers (Basel) 2024; 16:4141. [PMID: 39766041 PMCID: PMC11675003 DOI: 10.3390/cancers16244141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Gastric cancer (GC) is a malignant tumor of the gastrointestinal tract associated with high mortality rates and accounting for approximately 1 million new cases diagnosed annually. Surgery, particularly radical gastrectomy, remains the primary treatment; however, there are currently no specific approaches to better distinguish malignant from healthy tissue or to differentiate between metastatic and non-metastatic lymph nodes. As a result, surgeons have to remove all lymph nodes indiscriminately, increasing intraoperative risks for patients and prolonging hospital stay. Near-infrared fluorescence imaging with indocyanine green (ICG) can provide real-time visualization of the surgical field using both conventional laparoscopy and robotic mini-invasive precision surgery platforms. However, its application shows some limits, as ICG is a non-targeted contrast agent. Several studies are now investigating the potential efficacy of fluorescent targeted agents that could selectively bind to the tumor tissue, offering a valuable tool for metastatic mapping during robotic gastrectomy. This review aims to summarize the key fluorescent agents that have been developed to recognize GC markers, as well as those targeting the tumor microenvironment (TME) and metabolic features. These agents hold great potential as valuable tools for enhancing precision surgery in robotic gastrectomy procedures improving the clinical recovery of GC patients.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| | - Laura Fortuna
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| | - Francesco Coratti
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| | - Federico Passagnoli
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 50134 Florence, Italy
| | - Fabio Cianchi
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; (L.F.); (F.C.); (F.P.); (A.A.); (F.C.)
| |
Collapse
|
4
|
Huynh AS, Cohen AS, Doligalski M, Casagni TJ, Moberg VE, Huang X, Morse J, Abrahams D, Lloyd MC, Centeno BA, Baldwin MK, McLaughlin ML, Vagner J, Morse DL. Intraoperative Guidance of Pancreatic Cancer Resection Using a Toll-like Receptor 2-Targeted Fluorescence Molecular Imaging Agent. CANCER RESEARCH COMMUNICATIONS 2024; 4:2877-2887. [PMID: 39320054 PMCID: PMC11536076 DOI: 10.1158/2767-9764.crc-24-0244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/01/2024] [Accepted: 09/23/2024] [Indexed: 09/26/2024]
Abstract
To increase the achievement of negative R0 surgical margins and increase the low survival rates of pancreatic cancer, improvements in assessing tumor margins during surgical resections are needed. This can be accomplished by using pancreatic cancer-targeted fluorescence molecular imaging agents to intraoperatively detect tumor margins in real time. Because Toll-like receptor 2 (TLR2) is broadly expressed among many cancer types including pancreatic adenocarcinomas, a high-affinity TLR2-targeted fluorescence molecular imaging agent (TLR2L-800) was developed. We investigate the potential for increased survival by employing real-time intraoperative tumor detection in a preclinical orthotopic human pancreatic xenograft tumor model using TLR2L-800. Three cohorts of nude mice bearing orthotopic human pancreatic xenograft tumors were intravenously injected with TLR2L-800. At 24 hours postinjection, one cohort underwent in vivo fluorescence-guided surgical removal of tumors using a real-time fluorescence imaging platform, a second cohort underwent visible light surgery (VLS), and a third cohort did not undergo surgery. A fourth, nontumor-bearing cohort was administered TLR2L-800 with no surgery. At 41 days postsurgery, the survival rates were 53% for the fluorescence-guided surgery (FGS) group and 0% for both the VLS and the tumor-bearing no-surgery group. The overall 200-day survival rate of 35% for the FGS group was significant compared with 0% for the VLS group (P value = 0.0018). This study demonstrates the potential of increasing disease-free survival for patients with pancreatic cancer by increasing the attainment of R0 margins using a novel tumor-targeted lipopeptide ligand-based fluorescence molecular imaging agent, TLR2L-800, during real-time FGS. SIGNIFICANCE Human TLR2 is broadly expressed among pancreatic adenocarcinomas, and the highly specific TLR2L-800 fluorescence molecular imaging agent has potential for use in fluorescence-guided surgery to increase R0 margins and improve patient survival.
Collapse
Affiliation(s)
- Amanda S. Huynh
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Allison S. Cohen
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Michael Doligalski
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Todd J. Casagni
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Valerie E. Moberg
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Xuan Huang
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jennifer Morse
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Dominique Abrahams
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Mark C. Lloyd
- Analytic Microscopy Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Barbara A. Centeno
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Margaret K. Baldwin
- Department of Comparative Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Mark L. McLaughlin
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia
| | - Josef Vagner
- BIO5 Institute, University of Arizona, Tucson, Arizona
| | - David L. Morse
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Small Animal Imaging Laboratory, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Oncologic Sciences, University of South Florida, Tampa, Florida
- Department of Physics, University of South Florida, Tampa, Florida
- Department of Medical Engineering, University of South Florida, Tampa, Florida
| |
Collapse
|
5
|
Vaselli M, Gabriels RY, Schmidt I, Sterkenburg AJ, Kats-Ugurlu G, Nagengast WB, de Boer JF. Ex vivo optical coherence tomography combined with near infrared targeted fluorescence: towards in-vivo esophageal cancer detection. BIOMEDICAL OPTICS EXPRESS 2024; 15:5706-5722. [PMID: 39421768 PMCID: PMC11482167 DOI: 10.1364/boe.537828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 10/19/2024]
Abstract
Early detection of (pre)malignant esophageal lesions is critical to improve esophageal cancer morbidity and mortality rates. In patients with advanced esophageal adenocarcinoma (EAC) who undergo neoadjuvant chemoradiation therapy, the efficacy of therapy could be optimized and unnecessary surgery prevented by the reliable assessment of residual tumors after therapy. Optical coherence tomography (OCT) provides structural images at a (sub)-cellular level and has the potential to visualize morphological changes in tissue. However, OCT lacks molecular imaging contrast, a feature that enables the study of biological processes at a cellular level and can enhance esophageal cancer diagnostic accuracy. We combined OCT with near-infrared fluorescence molecular imaging using fluorescently labelled antibodies (immuno-OCT). The main goal of this proof of principle study is to investigate the feasibility of immuno-OCT for esophageal cancer imaging. We aim to assess whether the sensitivity of our immuno-OCT device is sufficient to detect the tracer uptake using an imaging dose (∼100 times smaller than a dose with therapeutic effects) of a targeted fluorescent agent. The feasibility of immuno-OCT was demonstrated ex-vivo on dysplastic lesions resected from Barrett's patients and on esophageal specimens resected from patients with advanced EAC, who were respectively topically and intravenously administrated with the tracer bevacizumab-800CW. The detection sensitivity of our system (0.3 nM) is sufficient to detect increased tracer uptake with micrometer resolution using an imaging dose of labelled antibodies. Moreover, the absence of layered structures that are typical of normal esophageal tissue observed in OCT images of dysplastic/malignant esophageal lesions may further aid their detection. Based on our preliminary results, immuno-OCT could improve the detection of dysplastic esophageal lesions.
Collapse
Affiliation(s)
- Margherita Vaselli
- Department of Physics and Astronomy, LaserLab Amsterdam, Vrije Universiteit de Boelelaan 1081,, Amsterdam, The Netherlands
| | - Ruben Y. Gabriels
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrea J. Sterkenburg
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gursah Kats-Ugurlu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wouter B. Nagengast
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johannes F. de Boer
- Department of Physics and Astronomy, LaserLab Amsterdam, Vrije Universiteit de Boelelaan 1081,, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Pan M, Zhao R, Fu C, Tang M, Zhou J, Ma B, Liu J, Yang Y, Chen B, Zhang Q, Wang Y. Tuning nanoparticle core composition drives orthogonal fluorescence amplification for enhanced tumour imaging. Nat Commun 2024; 15:7824. [PMID: 39242636 PMCID: PMC11379858 DOI: 10.1038/s41467-024-52029-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 08/23/2024] [Indexed: 09/09/2024] Open
Abstract
Tumour detection with high selectivity and sensitivity is crucial for delineating tumour margins and identifying metastatic foci during image-guided surgery. Optical nanoprobes with preferential tumour accumulation is often limited by inefficient amplification of biological signals. Here, we report the design of a library of hydrophobic core-tunable ultra-pH-sensitive nanoprobes (HUNPs) for orthogonally amplifying tumour microenvironmental signals on multiple tumour models. We find that tuning the hydrophobicity of nanoparticle core composition with non-ionizable monomers can enhance cellular association of HUNPs by more than ten-fold, resulting in a high cellular internalization efficiency of HUNPs with up to 50% in tumours. Combining high tumour accumulation and high cell internalization efficiency, HUNPs show orthogonally amplified fluorescence signals, permitting the precise locating and delineating margins between malignant lesions and normal tissues with high contrast-to-noise ratio and resolution. Our study provides key strategies to design nanomedicines with high intracellular bioavailability for cancer detection, drug/gene delivery, and therapy.
Collapse
Affiliation(s)
- Meijie Pan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Ruiyang Zhao
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Chuanxun Fu
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Mingmei Tang
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jiayi Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Bin Ma
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jianxiong Liu
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Ye Yang
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Binlong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Chemical Biology Center, Peking University, Beijing, China.
| |
Collapse
|
7
|
Zhao X, Du J, Sun W, Fan J, Peng X. Regulating Charge Transfer in Cyanine Dyes: A Universal Methodology for Enhancing Cancer Phototherapeutic Efficacy. Acc Chem Res 2024; 57:2582-2593. [PMID: 39152945 DOI: 10.1021/acs.accounts.4c00399] [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: 08/19/2024]
Abstract
Due to the advantages of spatiotemporal selectivity and inherent noninvasiveness, cancer phototherapy, which includes both photodynamic therapy (PDT) and photothermal therapy (PTT), has garnered significant attention in recent years as a promising cancer treatment. Despite the commendable progress in this field, persistent challenges remain. In PDT, limitations in dyes manifest as low intersystem crossing (ISC) efficiency and oxygen-dependent photoactivity, resulting in unsatisfactory performance, particularly under hypoxic conditions. Similarly, PTT encounters consistent insufficiencies in the photothermal conversion efficiency (PCE) of dyes. Additionally, the suboptimal phototherapeutic efficacy often exhibits a limited immune response. These factors collectively impose significant constraints on phototherapy in oncological applications, leading to limited tumor inhibition, tumor recurrence, and even metastasis. Unlike strategies that rely on external assistance with complicated systems, manipulating excited-state deactivation pathways in biocompatible dyes offers a universal way to systematically address these challenges. Our group has devoted considerable effort to achieving this goal. In this Account, we present and discuss our journey in optimizing excited-state energy-release pathways through regulating molecular charge transfer based on cyanine dyes, which are renowned for their exceptional photophysical properties and harmonious biocompatibility. The investigation begins with the introduction of amino groups in the meso position of a heptamethine cyanine dye, where the intramolecular charge transfer (ICT) effect causes a significant enlargement of the Stokes shift. Subsequently, ICT induced by introducing functional electron-deficient groups in cyanines is found to decrease the overlap of electron distribution or narrow the energy gaps of molecular frontier orbitals. Such modifications result in a reduction of the energy gaps between singlet and triplet states or an improvement in internal conversion, ultimately promoting phototherapy efficacy in both primary and distant tumors. Furthermore, with the intensification of the charge transfer effect aided by light, photoinduced intramolecular electron transfer occurs in some cyanines, leading to complete charge separation in the excited state. This process enhances the transition to the ground or triplet states, improving tumor phototherapy and inhibiting metastasis by increasing the PCE or the yield of reactive oxygen species, respectively. Shifting focus from intramolecular to intermolecular interactions, we successfully constructed and explored cyanines based on intermolecular charge transfer. These dyes, with excited-state dynamics mimicking natural photosynthesis, generate radicals and facilitate oxygen-independent hypoxic tumor PDT. Finally, we outlined the existing challenges and future directions for optimizing phototherapeutic efficacy by regulating molecular charge transfer. This Account provides molecular-level insights into improving phototherapeutic performance, offering valuable perspectives, and inspiring the development of functional dyes in other application fields.
Collapse
Affiliation(s)
- Xueze Zhao
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, 116024 Dalian, China
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, 116024 Dalian, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, 116024 Dalian, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, 116024 Dalian, China
- Ningbo Institute of Dalian University of Technology, No. 26 Yucai Road, 315016 Ningbo, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, No. 2 Linggong Road, 116024 Dalian, China
| |
Collapse
|
8
|
Zeng F, Li C, Wang H, Wang Y, Ren T, He F, Jiang J, Xu J, Wang B, Wu Y, Yu Y, Hu Z, Tian J, Wang S, Tang X. Intraoperative Resection Guidance and Rapid Pathological Diagnosis of Osteosarcoma using B7H3 Targeted Probe under NIR-II Fluorescence Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310167. [PMID: 38502871 PMCID: PMC11434027 DOI: 10.1002/advs.202310167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/31/2024] [Indexed: 03/21/2024]
Abstract
Complete removal of all tumor tissue with a wide surgical margin is essential for the treatment of osteosarcoma (OS). However, it's difficult, sometimes impossible, to achieve due to the invisible small satellite lesions and blurry tumor boundaries. Besides, intraoperative frozen-section analysis of resection margins of OS is often restricted by the hard tissues around OS, which makes it impossible to know whether a negative margin is achieved. Any unresected small tumor residuals will lead to local recurrence and worse prognosis. Herein, based on the high expression of B7H3 in OS, a targeted probe B7H3-IRDye800CW is synthesized by conjugating anti-B7H3 antibody and IRDye800CW. B7H3-IRDye800CW can accurately label OS areas after intravenous administration, thereby helping surgeons identify and resect residual OS lesions (<2 mm) and lung metastatic lesions. The tumor-background ratio reaches 4.42 ± 1.77 at day 3. After incubating fresh human OS specimen with B7H3-IRDye800CW, it can specifically label the OS area and even the microinvasion area (confirmed by hematoxylin-eosin [HE] staining). The probe labeled area is consistent with the tumor area shown by magnetic resonance imaging and complete HE staining of the specimen. In summary, B7H3-IRDye800CW has translational potential in intraoperative resection guidance and rapid pathological diagnosis of OS.
Collapse
Affiliation(s)
- Fanwei Zeng
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Changjian Li
- School of Engineering Medicine & Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, 100191, China
| | - Han Wang
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Yueqi Wang
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Fangzhou He
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Jie Jiang
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Jiuhui Xu
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Boyang Wang
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Yifan Wu
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Yiyang Yu
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jie Tian
- School of Engineering Medicine & Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, 100191, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Shidong Wang
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor & Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
9
|
Boland PA, Hardy NP, Moynihan A, McEntee PD, Loo C, Fenlon H, Cahill RA. Intraoperative near infrared functional imaging of rectal cancer using artificial intelligence methods - now and near future state of the art. Eur J Nucl Med Mol Imaging 2024; 51:3135-3148. [PMID: 38858280 PMCID: PMC11300525 DOI: 10.1007/s00259-024-06731-9] [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: 12/23/2023] [Accepted: 04/15/2024] [Indexed: 06/12/2024]
Abstract
Colorectal cancer remains a major cause of cancer death and morbidity worldwide. Surgery is a major treatment modality for primary and, increasingly, secondary curative therapy. However, with more patients being diagnosed with early stage and premalignant disease manifesting as large polyps, greater accuracy in diagnostic and therapeutic precision is needed right from the time of first endoscopic encounter. Rapid advancements in the field of artificial intelligence (AI), coupled with widespread availability of near infrared imaging (currently based around indocyanine green (ICG)) can enable colonoscopic tissue classification and prognostic stratification for significant polyps, in a similar manner to contemporary dynamic radiological perfusion imaging but with the advantage of being able to do so directly within interventional procedural time frames. It can provide an explainable method for immediate digital biopsies that could guide or even replace traditional forceps biopsies and provide guidance re margins (both areas where current practice is only approximately 80% accurate prior to definitive excision). Here, we discuss the concept and practice of AI enhanced ICG perfusion analysis for rectal cancer surgery while highlighting recent and essential near-future advancements. These include breakthrough developments in computer vision and time series analysis that allow for real-time quantification and classification of fluorescent perfusion signals of rectal cancer tissue intraoperatively that accurately distinguish between normal, benign, and malignant tissues in situ endoscopically, which are now undergoing international prospective validation (the Horizon Europe CLASSICA study). Next stage advancements may include detailed digital characterisation of small rectal malignancy based on intraoperative assessment of specific intratumoral fluorescent signal pattern. This could include T staging and intratumoral molecular process profiling (e.g. regarding angiogenesis, differentiation, inflammatory component, and tumour to stroma ratio) with the potential to accurately predict the microscopic local response to nonsurgical treatment enabling personalised therapy via decision support tools. Such advancements are also applicable to the next generation fluorophores and imaging agents currently emerging from clinical trials. In addition, by providing an understandable, applicable method for detailed tissue characterisation visually, such technology paves the way for acceptance of other AI methodology during surgery including, potentially, deep learning methods based on whole screen/video detailing.
Collapse
Affiliation(s)
- Patrick A Boland
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - N P Hardy
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - A Moynihan
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - P D McEntee
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland
| | - C Loo
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland
| | - H Fenlon
- Department of Radiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - R A Cahill
- UCD Centre for Precision Surgery, School of Medicine, University College Dublin, 47 Eccles Street, Dublin 7, Dublin, Ireland.
- Department of Colorectal Surgery, Mater Misericordiae University Hospital, Dublin, Ireland.
| |
Collapse
|
10
|
van Schaik JE, van der Vegt B, Slagter-Menkema L, Hanemaaijer SH, Halmos GB, Witjes MJH, van der Laan BFAM, Fehrmann RSN, Oosting SF, Plaat BEC. Potential imaging targets in primary head and neck squamous cell carcinoma and lymph node metastases. Am J Otolaryngol 2024; 45:104298. [PMID: 38640809 DOI: 10.1016/j.amjoto.2024.104298] [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: 04/04/2024] [Accepted: 04/14/2024] [Indexed: 04/21/2024]
Abstract
PURPOSE To investigate glycoprotein nonmetastatic melanoma protein B (GPNMB) and vascular endothelial growth factor (VEGF) as potential fluorescent imaging markers by comparing their protein expression to epidermal growth factor receptor (EGFR). MATERIALS AND METHODS Thirty-eight paired samples of untreated head and neck squamous cell carcinoma (HNSCC) primary tumours (PT) and corresponding synchronous lymph node metastases (LNM) were selected. After immunohistochemical staining, expression was assessed and compared by the percentage of positive tumour cells. Data were analysed using the Mann-Whitney test, effect sizes (ESr) and Spearman's correlation coefficient (r). RESULTS GPNMB expression was observed in 100 % of PT, and median 80 % (range 5-100 %) of tumour cells, VEGF in 92 % and 60 % (0-100 %), EGFR in 87 % and 60 % (0-100 %) respectively. In corresponding LNM, GPNMB expression was observed in 100 % of LNM and median 90 % (20-100 %) of tumour cells, VEGF in 87 % and 65 % (0-100 %), and EGFR in 84 % and 35 % (0-100 %). A positive correlation was found between expression in PT and LNM for GPNMB (r = 0.548) and EGFR (r = 0.618) (p < 0.001), but not for VEGF (r = -0.020; p = 0.905). GPNMB expression was present in a higher percentage of tumour cells compared to EGFR in PT (p = 0.015, ESr = -0.320) and in LNM (p < 0.001, ESr = -0.478), while VEGF was not (p = 1.00, ESr = -0.109 and - 0.152, respectively). CONCLUSION GPNMB expression is higher than EGFR in untreated HNSCC PT and corresponding LNM, while VEGF expression is comparable to EGFR. GPNMB is a promising target for fluorescent imaging in HNSCC.
Collapse
Affiliation(s)
- Jeroen E van Schaik
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Lorian Slagter-Menkema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Saskia H Hanemaaijer
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Gyorgi B Halmos
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Max J H Witjes
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Bernard F A M van der Laan
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Sjoukje F Oosting
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Boudewijn E C Plaat
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands.
| |
Collapse
|
11
|
Houvast RD, Badr N, March T, de Muynck LDAN, Sier VQ, Schomann T, Bhairosingh S, Baart VM, Peeters JAHM, van Westen GJP, Plückthun A, Burggraaf J, Kuppen PJK, Vahrmeijer AL, Sier CFM. Preclinical evaluation of EpCAM-binding designed ankyrin repeat proteins (DARPins) as targeting moieties for bimodal near-infrared fluorescence and photoacoustic imaging of cancer. Eur J Nucl Med Mol Imaging 2024; 51:2179-2192. [PMID: 37642704 PMCID: PMC11178671 DOI: 10.1007/s00259-023-06407-w] [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: 05/27/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE Fluorescence-guided surgery (FGS) can play a key role in improving radical resection rates by assisting surgeons to gain adequate visualization of malignant tissue intraoperatively. Designed ankyrin repeat proteins (DARPins) possess optimal pharmacokinetic and other properties for in vivo imaging. This study aims to evaluate the preclinical potential of epithelial cell adhesion molecule (EpCAM)-binding DARPins as targeting moieties for near-infrared fluorescence (NIRF) and photoacoustic (PA) imaging of cancer. METHODS EpCAM-binding DARPins Ac2, Ec4.1, and non-binding control DARPin Off7 were conjugated to IRDye 800CW and their binding efficacy was evaluated on EpCAM-positive HT-29 and EpCAM-negative COLO-320 human colon cancer cell lines. Thereafter, NIRF and PA imaging of all three conjugates were performed in HT-29_luc2 tumor-bearing mice. At 24 h post-injection, tumors and organs were resected and tracer biodistributions were analyzed. RESULTS Ac2-800CW and Ec4.1-800CW specifically bound to HT-29 cells, but not to COLO-320 cells. Next, 6 nmol and 24 h were established as the optimal in vivo dose and imaging time point for both DARPin tracers. At 24 h post-injection, mean tumor-to-background ratios of 2.60 ± 0.3 and 3.1 ± 0.3 were observed for Ac2-800CW and Ec4.1-800CW, respectively, allowing clear tumor delineation using the clinical Artemis NIRF imager. Biodistribution analyses in non-neoplastic tissue solely showed high fluorescence signal in the liver and kidney, which reflects the clearance of the DARPin tracers. CONCLUSION Our encouraging results show that EpCAM-binding DARPins are a promising class of targeting moieties for pan-carcinoma targeting, providing clear tumor delineation at 24 h post-injection. The work described provides the preclinical foundation for DARPin-based bimodal NIRF/PA imaging of cancer.
Collapse
Affiliation(s)
- Ruben D Houvast
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.
| | - Nada Badr
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Taryn March
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Vincent Q Sier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Timo Schomann
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Shadhvi Bhairosingh
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Victor M Baart
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith A H M Peeters
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Gerard J P van Westen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Andreas Plückthun
- Department of Biochemistry, University of Zürich, Zurich, Switzerland
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Centre for Human Drug Research, Leiden, the Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
12
|
Chen S, Li G, Pan R, Zhou K, Wen W, Tao J, Wang F, Han RPS, Pan H, Tu Y. Novel Near-Infrared Fluorescent Probe for Hepatocyte Growth Factor in Vivo Imaging in Surgical Navigation of Colorectal Cancer. Anal Chem 2024; 96:9016-9025. [PMID: 38780636 DOI: 10.1021/acs.analchem.4c00350] [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: 05/25/2024]
Abstract
Despite recent advancements in colorectal cancer (CRC) treatment, the prognosis remains unfavorable primarily due to high recurrence and liver metastasis rates. Fluorescence molecular imaging technologies, combined with specific probes, have gained prominence in facilitating real-time tumor resection guided by fluorescence. Hepatocyte growth factor (HGF) is overexpressed in CRC, but the advancement of HGF fluorescent probes has been impeded by the absence of effective HGF-targeting small-molecular ligands. Herein, we present the targeted capabilities of the novel V-1-GGGK-MPA probe labeled with a near-infrared fluorescent dye, which targets HGF in CRC. The V-1-GGGK peptide exhibits high specificity and selectivity for HGF-positive in vitro tumor cells and in vivo tumors. Biodistribution analysis of V-1-GGGK-MPA revealed tumor-specific accumulation with low background uptake, yielding signal-to-noise ratio (SNR) values of tumor-to-colorectal >6 in multiple subcutaneous CRC models 12 h postinjection. Quantitative analysis confirmed the probe's high uptake in SW480 and HT29 orthotopic and liver metastatic models, with SNR values of tumor-to-colorectal and -liver being 5.6 ± 0.4, 4.6 ± 0.5, and 2.1 ± 0.3, 2.0 ± 0.5, respectively, enabling precise tumor visualization for surgical navigation. Pathological analysis demonstrated the excellent tumor boundaries discrimination capacity of the V-1-GGGK-MPA probe at the molecular level. With its rapid tumor targeting, sustained tumor retention, and precise tumor boundary delineation, V-1-GGGK-MPA merges as a promising HGF imaging agent, enriching the toolbox of intraoperative navigational fluorescent probes for CRC.
Collapse
Affiliation(s)
- Shuying Chen
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Gang Li
- Department of Ecology and Environment, Yuzhang Normal University, Nanchang 330103, China
| | - Rongbin Pan
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Kuncheng Zhou
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Weijie Wen
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ji Tao
- Human Phenome Institute, Fudan University, Shanghai 201203, China
| | - Fang Wang
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ray P S Han
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Huaping Pan
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yuanbiao Tu
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| |
Collapse
|
13
|
Zhou K, Li G, Pan R, Xin S, Wen W, Wang H, Luo C, Han RPS, Gu Y, Tu Y. Preclinical evaluation of AGTR1-Targeting molecular probe for colorectal cancer imaging in orthotopic and liver metastasis mouse models. Eur J Med Chem 2024; 271:116452. [PMID: 38685142 DOI: 10.1016/j.ejmech.2024.116452] [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: 11/15/2023] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
Despite advancements in colorectal cancer (CRC) treatment, the prognosis remains unfavorable for patients with distant liver metastasis. Fluorescence molecular imaging with specific probes is increasingly used to guide CRC surgical resection in real-time and treatment planning. Here, we demonstrate the targeted imaging capacity of an MPA-PEG4-N3-Ang II probe labeled with near-infrared (NIR) fluorescent dye targeting the angiotensin II (Ang II) type 1 receptor (AGTR1) that is significantly upregulated in CRC. MPA-PEG4-N3-Ang II was highly selective and specific to in vitro tumor cells and in vivo tumors in a mouse CRC xenograft model. The favorable ex vivo imaging and in vivo biodistribution of MPA-PEG4-N3-Ang II afforded tumor-specific accumulation with low background and >10 contrast tumor-to-colorectal values in multiple subcutaneous CRC models at 8 h following injection. Biodistribution analysis confirmed the probe's high uptake in HT29 and HCT116 orthotopic and liver metastatic models of CRC with signal-to-noise ratio (SNR) values of tumor-to-colorectal and -liver fluorescence of 5.8 ± 0.6, 5.3 ± 0.7, and 2.7 ± 0.5, 2.6 ± 0.5, respectively, enabling high-contrast intraoperative tumor visualization for surgical navigation. Given its rapid tumor targeting, precise tumor boundary delineation, durable tumor retention and docking study, MPA-PEG4-N3-Ang II is a promising high-contrast imaging agent for the clinical detection of CRC.
Collapse
Affiliation(s)
- Kuncheng Zhou
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Gang Li
- Department of Ecology and Environment, Yuzhang Normal University, Nanchang, 330103, China
| | - Rongbin Pan
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Sulin Xin
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Weijie Wen
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Huiyi Wang
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Chao Luo
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Ray P S Han
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yueqing Gu
- State Key Laboratory of Natural Medicine, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yuanbiao Tu
- Cancer Research Center, the Jiangxi Province Key Laboratory for Diagnosis, Treatment, and Rehabilitation of Cancer in Chinese Medicine, Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| |
Collapse
|
14
|
Shi Q, Xu J, Xu H, Wang Q, Huang S, Wang X, Wang P, Hu F. Polystyrene-Based Matrix to Enhance the Fluorescence of Aggregation-Induced Emission Luminogen for Fluorescence-Guided Surgery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309589. [PMID: 38105589 DOI: 10.1002/smll.202309589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/13/2023] [Indexed: 12/19/2023]
Abstract
Achieving ultrabright fluorogens is a key issue for fluorescence-guided surgery (FGS). Fluorogens with aggregation-induced emission (AIEgens) are potential agents for FGS on the benefit of the bright fluorescence in physiological conditions. Herein, the fluorescence brightness of AIEgen is further improved by preparing the nanoparticle using a polystyrene-based matrix and utilizing it for tumor FGS with a high signal-to-background ratio. After encapsulating AIEgen into polystyrene-poly (ethylene glycol) (PS-PEG), the fluorescence intensity of the prepared AIE@PS-PEG nanoparticles is multiple times that of nanoparticles in 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-poly (ethylene glycol) (DSPE-PEG), a commonly used polymer matrix for nanoparticle preparation. Molecular dynamics simulations suggest that higher free energy is required for the outer rings of AIEgen to rotate in polystyrene than in the DSPE, indicating that the benzene rings in polystyrene can restrict the intramolecular motions of AIEgen better than the alkyl chain in DSPE-PEG. Fluorescence correlation microscopy detections suggest that the triplet excited state of AIEgens is less in PS-PEG than in DSPE-PEG. The restricted intramolecular motions and suppressed triplet excited state result in ultrabright AIE@PS-PEG nanoparticles, which are more conducive to illuminating tumor tissues in the intestine for FGS. The illumination of metastatic tumors in lungs by AIE@PS-PEG nanoparticles is also tried.
Collapse
Affiliation(s)
- Qiankun Shi
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jieying Xu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Huihui Xu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Qiang Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Shaohui Huang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 101408, China
| | - Xiaorui Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Peng Wang
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Fang Hu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| |
Collapse
|
15
|
Rijsemus CJV, Kok NFM, Aalbers AGJ, Grotenhuis BA, Berardi E, Snaebjornsson P, Lambregts DMJ, Beets-Tan RGH, Lahaye MJ. Investigating locations of recurrences with MRI after CRS-HIPEC for colorectal peritoneal metastases. Eur J Radiol 2024; 175:111478. [PMID: 38677041 DOI: 10.1016/j.ejrad.2024.111478] [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: 10/08/2023] [Revised: 03/13/2024] [Accepted: 04/21/2024] [Indexed: 04/29/2024]
Abstract
PURPOSE Patients with colorectal peritoneal metastases (PM) treated with cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are at high risk of recurrent disease. Understanding where and why recurrences occur is the first step in finding solutions to reduce recurrence rates. Although diffusion-weighted (DW) MRI is not routinely used in the follow-up of CRC patients, it has a clear advantage over CT in detecting the location and spread of (recurrent) PM. This study aimed to identify common locations of recurrence in CRC patients after CRS-HIPEC with MRI. METHOD This was a single-centre retrospective study of patients with recurrent PM after CRS-HIPEC performed between January 2016 and August 2020. Patients were eligible for inclusion if they had both an MRI preoperatively (MRI1) and at the time of recurrent disease (MRI2). Two abdominal radiologists reviewed in consensus and categorized recurrences according to their location on MRI2 and in correlation with previous disease location on prior imaging (MRI1) and the surgical report of the CRS-HIPEC. RESULTS Thirty patients were included, with a median surgical PCI of 7 (range 3-21) at the time of primary CRS-HIPEC. In total, 68 recurrent metastases were detected on MRI2, of which 14 were extra-peritoneal. Of the remaining 54 PM, 42 (78%) occurred where the peritoneum was damaged due to earlier resections or other surgical procedures (e.g. inserted surgical abdominal drains). Most recurrent metastases were found in the mesentery, lower abdomen/pelvis and abdominal wall (87%). CONCLUSIONS Most recurrent PMs appeared in the mesentery, lower abdomen/pelvis and abdominal wall, especially where the peritoneum was previously damaged.
Collapse
Affiliation(s)
- C J V Rijsemus
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands; Department of Surgery, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands; GROW School for Oncology and Developmental Biology - University of Maastricht, Maastricht, the Netherlands.
| | - N F M Kok
- Department of Surgery, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands
| | - A G J Aalbers
- Department of Surgery, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands
| | - B A Grotenhuis
- Department of Surgery, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands
| | - E Berardi
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands
| | - P Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands; Department of Pathology, Faculty of Medicine - University of Iceland, Reykjavik, Iceland
| | - D M J Lambregts
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands
| | - R G H Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands; GROW School for Oncology and Developmental Biology - University of Maastricht, Maastricht, the Netherlands
| | - M J Lahaye
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121 1066CX, Amsterdam, the Netherlands
| |
Collapse
|
16
|
Lin Q, Li C, Wang Y, Zhu Y, Gu Y. Discovery of Near-Infrared Heptamethine Cyanine Probes for Imaging-Guided Surgery in Solid Tumors. J Med Chem 2024; 67:5800-5812. [PMID: 38560986 DOI: 10.1021/acs.jmedchem.4c00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Near-infrared (NIR) fluorescence imaging has attracted much attention in image-guided interventions with unique advantages. However, the clinical translation rate of fluorescence probes is extremely low, primarily due to weak lesion signal contrast and poor specificity. To address this dilemma, a series of small-molecule near-infrared fluorescence probes have been designed for tumor imaging. Among them, YQ-04-03 showed notable optical stability and remarkable sensitivity toward tumor targeting. Moreover, within a specific concentration and time range against oxidizing reducing agents and laser, it demonstrated better stability than ICG. The retention time of YQ-04-03 in tumors was significantly longer compared to other nonspecific uptake sites in the subjects, and its tumor-to-normal tissue ratio (TNR) outperformed ICG. Successful resection of in situ hepatocarcinoma and peritoneal carcinoma was achieved using probe imaging guidance, with the smallest visual lesion resected measuring approximately 1 mm3. Ultimately, this probe holds great potential for advancing tumor tracer.
Collapse
Affiliation(s)
- Qiao Lin
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Changsheng Li
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
- Nanjing Nuoyuan Medical Devices Co., Ltd, NO.18 Ziyun Avenue, Qinhuai District, Nanjing 210000, China
| | - Yuhua Wang
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yanqing Zhu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
17
|
Peters I, Marchetti C, Scambia G, Fagotti A. New windows of surgical opportunity for gynecological cancers in the era of targeted therapies. Int J Gynecol Cancer 2024; 34:352-362. [PMID: 38438181 DOI: 10.1136/ijgc-2023-004580] [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: 03/06/2024] Open
Abstract
Precision medicine through molecular profiling has taken a prominent role in the treatment of solid tumors and it is widely expected that this will continue to expand. With respect to gynecological cancers, a major change has particularly been observed in the treatment landscape of epithelial ovarian, endometrial, and cervical cancers. Regarding the former, maintenance therapy with either poly(ADP-ribose) polymerase inhibitors (PARPi) and/or bevacizumab has become an indispensable treatment option following the traditional combination of cytoreductive surgery and platinum-based chemotherapy. Considering endometrial cancer, the molecular classification system has now been incorporated into virtually every guideline available and molecular-directed treatment strategies are currently being researched, presumably leading to a further transformation of its treatment paradigm. After all, treatment with immune-checkpoint inhibitors that target the programmed cell death 1 (PD-1) receptor has already been shown to significantly improve disease outcomes in these patients, especially in those with mismatch repair deficient, microsatellite stability-high (MMRd-MSI-H) disease. Similarly, in recurrent/metastatic cervical cancer patients, these agents elicited improved survival rates when being added to platinum-based chemotherapy with or without bevacizumab. Interestingly, implications of these targeted therapies for surgical management have been touched on to a minor extent, but are at least as intriguing. This review therefore aims to address the wide-ranging opportunities the molecular tumor characteristics and their corresponding targeted therapies have to offer for the surgical management of epithelial ovarian, endometrial, and cervical cancers, both in the primary and recurrent setting.
Collapse
Affiliation(s)
- Inge Peters
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Claudia Marchetti
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Giovanni Scambia
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Anna Fagotti
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
18
|
Giuliani S, Paraboschi I, McNair A, Smith M, Rankin KS, Elson DS, Paleri V, Leff D, Stasiuk G, Anderson J. Monoclonal Antibodies for Targeted Fluorescence-Guided Surgery: A Review of Applicability across Multiple Solid Tumors. Cancers (Basel) 2024; 16:1045. [PMID: 38473402 PMCID: PMC10931077 DOI: 10.3390/cancers16051045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
This study aims to review the status of the clinical use of monoclonal antibodies (mAbs) that have completed or are in ongoing clinical trials for targeted fluorescence-guided surgery (T-FGS) for the intraoperative identification of the tumor margins of extra-hematological solid tumors. For each of them, the targeted antigen, the mAb generic/commercial name and format, and clinical indications are presented, together with utility, doses, and the timing of administration. Based on the current scientific evidence in humans, the top three mAbs that could be prepared in a GMP-compliant bank ready to be delivered for surgical purposes are proposed to speed up the translation to the operating room and produce a few readily available "off-the-shelf" injectable fluorescent probes for safer and more effective solid tumor resection.
Collapse
Affiliation(s)
- Stefano Giuliani
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, University College London, London W1W 7TY, UK
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK;
- Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Trust, London WC1N 3JH, UK
| | - Irene Paraboschi
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milan, Italy;
| | - Angus McNair
- National Institute for Health Research Bristol Biomedical Research Centre, Bristol Centre for Surgical Research, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK;
- Department of Gastrointestinal Surgery, North Bristol NHS Trust, Bristol BS10 5NB, UK
| | - Myles Smith
- The Sarcoma, Melanoma and Rare Tumours Unit, The Royal Marsden Hospital, Institute Cancer of Research, London SW3 6JJ, UK;
| | - Kenneth S. Rankin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| | - Daniel S. Elson
- Hamlyn Centre for Robotic Surgery, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK;
| | - Vinidh Paleri
- Head and Neck Unit, The Royal Marsden Hospitals, London SW3 6JJ, UK;
| | - Daniel Leff
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK;
| | - Graeme Stasiuk
- Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King’s College London, St Thomas’ Hospital, London SE1 7EH, UK;
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK;
| |
Collapse
|
19
|
Kondo T, Nishio N, Park JS, Mani LD, Naveed A, Tanaka H, Lewis JS, Rosenthal EL, Hom ME. Identification of Optimal Tissue-Marking Dye Color for Pathological Evaluation in Fluorescence Imaging Using IRDye800CW. Mol Imaging Biol 2024; 26:162-172. [PMID: 38057647 DOI: 10.1007/s11307-023-01882-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
PURPOSE Fluorescence-guided surgery using a tumor-specific antibody-dye conjugate is useful in various cancer types. Fluorescence imaging is a valuable tool both intraoperatively and postoperatively for ex vivo imaging. The color of inks used for tumor specimens during ex vivo specimen processing in pathology is an important consideration for fluorescence imaging since the absorption/emission of the dyes may interfere with the fluorescent dye. This study assesses suitable ink colors for use specifically with IRDye800CW fluorescence imaging. PROCEDURES Eight tissue-marking inks or dyes (TMDs) commonly used for pathological evaluation were assessed. Agarose tissue-mimicking phantoms containing Panitumumab-IRDye800CW were used as an initial model. Mean fluorescence intensity was measured at 800 nm using both Pearl Trilogy as a closed-field fluorescence imaging system and pde-neo II as an open-field fluorescence imaging system before and after TMD application. An in vivo mouse xenograft model using the human head and neck squamous cell carcinoma FaDu cell line was then used in conjunction with TMDs. RESULTS The retained IRDye800CW fluorescence on Pearl Trilogy was as follows: yellow at 91.0 ± 4.5%, red at 90.6 ± 2.7%, orange at 88.2 ± 2.2%, violet at 56.6 ± 1.1%, lime at 40.9 ± 1.8%, green at 19.3 ± 2.8%, black at 13.3 ± 0.6%, and blue at 8.1 ± 0.2%. The retained IRDye800CW fluorescence on pde-neo II was as follows: yellow at 86.5 ± 6.4%, red at 77.0 ± 6.2%, orange at 76.9 ± 2.8%, lime at 72.5 ± 9.5%, violet at 59.7 ± 0.4%, green at 30.1 ± 6.9%, black at 17.0 ± 2.7%, and blue at 6.7 ± 1.7%. The retained IRDye800CW fluorescence in yellow and blue TMDs was 42.1 ± 14.9% and 0.2 ± 0.2%, respectively in the mouse experiment (p = 0.039). CONCLUSION Yellow, red, and orange TMDs should be used, and blue and black TMDs should be avoided for evaluating tumor specimens through fluorescence imaging using IRDye800CW.
Collapse
Affiliation(s)
- Takahito Kondo
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naoki Nishio
- Department of Otorhinolaryngology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Jason S Park
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lucas D Mani
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Abdullah Naveed
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hidenori Tanaka
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James S Lewis
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eben L Rosenthal
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marisa E Hom
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
Shevchenko I, Serban D, Dascalu AM, Tribus L, Alius C, Cristea BM, Suceveanu AI, Voiculescu D, Dumitrescu D, Bobirca F, Suceveanu AP, Georgescu DE, Serboiu CS. Factors Affecting the Efficiency of Near-Infrared Indocyanine Green (NIR/ICG) in Lymphatic Mapping for Colorectal Cancer: A Systematic Review. Cureus 2024; 16:e55290. [PMID: 38558607 PMCID: PMC10981778 DOI: 10.7759/cureus.55290] [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] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
As laparoscopy gained global popularity in oncologic surgery, the challenge of detecting lymph nodes spurred researchers to explore innovative techniques and approach the situation from a fresh perspective. While many proposed methods have faded into obscurity, the utilization of indocyanine green (ICG) in the surgical treatment of oncologic patients has continued to advance. The immense potential of this dye is widely acknowledged, yet its full extent and limitations in lymphatic mapping for colorectal cancer remain to be precisely determined. This article aims to assess the magnitude of its potential and explore the constraints based on insights from clinical studies published by pioneering researchers. A systematic review of the existing literature, comprising articles in English, was conducted using the Scopus, PubMed, and Springer Link databases. The search employed keywords such as "colorectal cancer" AND/OR "indocyanine green," "fluorescence" AND/OR "lymphatic mapping" AND/OR "lymph nodes." Initially identifying 129 articles, the application of selection criteria narrowed down the pool to 10 articles, which served as the primary sources of data for our review. Despite the absence of a standardized protocol for the application of ICG in colorectal cancer, particularly in the context of lymphatic mapping, the detection rates have exhibited considerable variation across studies. Nevertheless, all authors unanimously regarded this technique as beneficial and promising. Additionally, it is advocated as an adjunctive tool to enhance the accuracy of cancer staging. Near-infrared (NIR)-enhanced surgery holds the promise of transforming the landscape of oncologic surgery, emerging as a valuable tool for surgeons. However, the absence of a standardized technique and the subjective nature of result assessment impose limitations on the potential of this method. Consequently, it can be inferred that the establishment of a universally accepted protocol, encompassing parameters such as dose, concentration, technique, and site of administration of ICG, along with the optimal time needed for fluorescence visualization, would enhance the outcomes. Emphasizing the accurate selection of patients is crucial to prevent the occurrence of false-negative results.
Collapse
Affiliation(s)
- Irina Shevchenko
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Dragos Serban
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Ana Maria Dascalu
- Ophthalmology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Laura Tribus
- Gastroenterology, Faculty of Oral Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Catalin Alius
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Bogdan Mihai Cristea
- Anatomy, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Andra Iulia Suceveanu
- Gastroenterology, Faculty of Medicine, Ovidius University of Constanta, Constanta, ROU
- Gastroenterology, Clinical Emergency Hospital St Apostle Andrew, Constanta, ROU
| | - Daniel Voiculescu
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Dan Dumitrescu
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Emergency University Hospital Bucharest, Bucharest, ROU
| | - Florin Bobirca
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
- General Surgery, Clinic Hospital "Dr. Ioan Cantacuzino" Bucharest, Bucharest, ROU
| | - Adrian Paul Suceveanu
- Medicine, Faculty of Medicine, Ovidius University of Constanta, Constanta, ROU
- Gastroenterology, Clinical Emergency Hospital St Apostle Andrew, Constanta, ROU
| | - Dragos Eugen Georgescu
- Surgery, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| | - Crenguta Sorina Serboiu
- Radiology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, ROU
| |
Collapse
|
21
|
Rounds CC, de Wit JG, Vonk J, Vorjohan J, Nelson S, Trang A, Villinski B, Samkoe KS, Brankov JG, Voskuil FJ, Witjes MJH, Tichauer KM. Improved intraoperative identification of close margins in oral squamous cell carcinoma resections using a dual aperture fluorescence ratio approach: first in-human results. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:016003. [PMID: 38235321 PMCID: PMC10793906 DOI: 10.1117/1.jbo.29.1.016003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024]
Abstract
Significance Surgical excision is the main treatment for solid tumors in oral squamous cell carcinomas, where wide local excision (achieving a healthy tissue margin of > 5 mm around the excised tumor) is the goal as it results in reduced local recurrence rates and improved overall survival. Aim No clinical methods are available to assess the complete surgical margin intraoperatively while the patient is still on the operating table; and while recent intraoperative back-bench fluorescence-guided surgery approaches have shown promise for detecting "positive" inadequate margins (< 1 mm ), they have had limited success in the detection of "close" inadequate margins (1 to 5 mm). Here, a dual aperture fluorescence ratio (dAFR) approach was evaluated as a means of improving detection of close margins. Approach The approach was evaluated on surgical specimens from patients who were administered a tumor-specific fluorescent imaging agent (cetuximab-800CW) prior to surgery. The dAFR approach was compared directly against standard wide-field fluorescence imaging and pathology measurements of margin thickness in specimens from three patients and a total of 12 margin locations (1 positive, 5 close, and 6 clear margins). Results The area under the receiver operating characteristic curve, representing the ability to detect close compared to clear margins (> 5 mm ) was found to be 1.0 and 0.57 for dAFR and sAF, respectively. Improvements in dAFR were found to be statistically significant (p < 0.02 ). Conclusions These results provide evidence that the dAFR approach potentially improves detection of close surgical margins.
Collapse
Affiliation(s)
- Cody C. Rounds
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Jaron G. de Wit
- University Medical Center Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Jasper Vonk
- University Medical Center Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
- University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
| | - Jennifer Vorjohan
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Sophia Nelson
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Allyson Trang
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Brooke Villinski
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| | - Kimberley S. Samkoe
- Dartmouth College, Thayer School of Engineering, Hanover, New Hampshire, United States
| | - Jovan G. Brankov
- University Medical Center Groningen, Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, Groningen, The Netherlands
- Illinois Institute of Technology, Department of Electrical and Computer Engineering, Chicago Illinois, United States
| | - Floris J. Voskuil
- University Medical Center Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Max J. H. Witjes
- University Medical Center Groningen, Department of Oral and Maxillofacial Surgery, Groningen, The Netherlands
| | - Kenneth M. Tichauer
- Illinois Institute of Technology, Department of Biomedical Engineering, Chicago, Illinois, United States
| |
Collapse
|
22
|
Sikkenk DJ, Sterkenburg AJ, Burghgraef TA, Akol H, Schwartz MP, Arensman R, Verheijen PM, Nagengast WB, Consten ECJ. Robot-assisted fluorescent sentinel lymph node identification in early-stage colon cancer. Surg Endosc 2023; 37:8394-8403. [PMID: 37721591 PMCID: PMC10615938 DOI: 10.1007/s00464-023-10394-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/13/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Patients with cT1-2 colon cancer (CC) have a 10-20% risk of lymph node metastases. Sentinel lymph node identification (SLNi) could improve staging and reduce morbidity in future organ-preserving CC surgery. This pilot study aimed to assess safety and feasibility of robot-assisted fluorescence-guided SLNi using submucosally injected indocyanine green (ICG) in patients with cT1-2N0M0 CC. METHODS Ten consecutive patients with cT1-2N0M0 CC were included in this prospective feasibility study. Intraoperative submucosal, peritumoral injection of ICG was performed during a colonoscopy. Subsequently, the near-infrared fluorescence 'Firefly' mode of the da Vinci Xi robotic surgical system was used for SLNi. SLNs were marked with a suture, after which a segmental colectomy was performed. The SLN was postoperatively ultrastaged using serial slicing and immunohistochemistry, in addition to the standard pathological examination of the specimen. Colonoscopy time, detection time (time from ICG injection to first SLNi), and total SLNi time were measured (time from the start of colonoscopy to start of segmental resection). Intraoperative, postoperative, and pathological outcomes were registered. RESULTS In all patients, at least one SLN was identified (mean 2.3 SLNs, SLN diameter range 1-13 mm). No tracer-related adverse events were noted. Median colonoscopy time was 12 min, detection time was 6 min, and total SLNi time was 30.5 min. Two patients had lymph node metastases present in the SLN, and there were no patients with false negative SLNs. No patient was upstaged due to ultrastaging of the SLN after an initial negative standard pathological examination. Half of the patients unexpectedly had pT3 tumours. CONCLUSIONS Robot-assisted fluorescence-guided SLNi using submucosally injected ICG in ten patients with cT1-2N0M0 CC was safe and feasible. SLNi was performed in an acceptable timespan and SLNs down to 1 mm were detected. All lymph node metastases would have been detected if SLN biopsy had been performed.
Collapse
Affiliation(s)
- Daan J Sikkenk
- Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
- Department of Surgery, Meander Medical Center, Maatweg 3, 3813 TZ, Amersfoort, The Netherlands
| | - Andrea J Sterkenburg
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Thijs A Burghgraef
- Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
- Department of Surgery, Meander Medical Center, Maatweg 3, 3813 TZ, Amersfoort, The Netherlands
| | - Halil Akol
- Department of Gastroenterology, Meander Medical Center, Maatweg 3, 3813 TZ, Amersfoort, The Netherlands
| | - Matthijs P Schwartz
- Department of Gastroenterology, Meander Medical Center, Maatweg 3, 3813 TZ, Amersfoort, The Netherlands
| | - René Arensman
- Department of Pathology, Meander Medical Center, Maatweg 3, 3813 TZ, Amersfoort, The Netherlands
| | - Paul M Verheijen
- Department of Surgery, Meander Medical Center, Maatweg 3, 3813 TZ, Amersfoort, The Netherlands
| | - Wouter B Nagengast
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Esther C J Consten
- Department of Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
- Department of Surgery, Meander Medical Center, Maatweg 3, 3813 TZ, Amersfoort, The Netherlands.
| |
Collapse
|
23
|
Ueda K, Ushijima H, Kawamura J. Lymphatic flow mapping during colon cancer surgery using indocyanine green fluorescence imaging. MINIM INVASIV THER 2023; 32:233-239. [PMID: 36628437 DOI: 10.1080/13645706.2022.2164468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/29/2022] [Indexed: 01/12/2023]
Abstract
With the development of surgical technology, indocyanine green (ICG) fluorescence navigation systems may be useful in various areas of colorectal surgery, including tumor location confirmation, bowel perfusion, ureter identification, and lymph node mapping. This review provides an overview of the current status of ICG-based navigation surgery in colorectal surgery, emphasizing its role in lymphatic flow mapping. This state-of-the-art approach will allow for appropriate oncological surgeries in the field of colorectal cancer and improve the patient's prognosis.
Collapse
Affiliation(s)
- Kazuki Ueda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka Sayama, Japan
| | - Hokuto Ushijima
- Department of Surgery, Kindai University Faculty of Medicine, Osaka Sayama, Japan
| | - Junichiro Kawamura
- Department of Surgery, Kindai University Faculty of Medicine, Osaka Sayama, Japan
| |
Collapse
|
24
|
Qiu Y, Yuan B, Cao Y, He X, Akakuru OU, Lu L, Chen N, Xu M, Wu A, Li J. Recent progress on near-infrared fluorescence heptamethine cyanine dye-based molecules and nanoparticles for tumor imaging and treatment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1910. [PMID: 37305979 DOI: 10.1002/wnan.1910] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023]
Abstract
Recenly, near-infrared fluorescence heptamethine cyanine dyes have shown satisfactory values in bioengineering, biology, and pharmacy especially in cancer diagnosis and treatment, owing to their excellent fluorescence property and biocompatibility. In order to achieve broad application prospects, diverse structures, and chemical properties of heptamethine cyanine dyes have been designed to develop novel functional molecules and nanoparticles over the past decade. For fluorescence and photoacoustic tumor imaging properties, heptamethine cyanine dyes are equipped with good photothermal performance and reactive oxygen species production properties under near-infrared light irradiation, thus holding great promise in photodynamic and/or photothermal cancer therapies. This review offers a comprehensive scope of the structures, comparisons, and applications of heptamethine cyanine dyes-based molecules as well as nanoparticles in tumor treatment and imaging in current years. Therefore, this review may drive the development and innovation of heptamethine cyanine dyes, significantly offering opportunities for improving tumor imaging and treatment in a precise noninvasive manner. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yue Qiu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Bo Yuan
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Yi Cao
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuelu He
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Ozioma Udochukwu Akakuru
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Liheng Lu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Nengwen Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Mengting Xu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Aiguo Wu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, Guangdong, China
| | - Juan Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Zhejiang International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Ningbo Cixi Institute of Biomedical Engineering, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, Guangdong, China
| |
Collapse
|
25
|
Qin X, Su M, Guo H, Peng B, Luo R, Ye J, Wang H. Functional biomaterials for the diagnosis and treatment of peritoneal surface malignancies. SMART MEDICINE 2023; 2:e20230013. [PMID: 39188342 PMCID: PMC11235712 DOI: 10.1002/smmd.20230013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/03/2023] [Indexed: 08/28/2024]
Abstract
Peritoneal surface malignancies (PSM) can originate from tumors in many organs and are highly malignant, and difficult to diagnose and cure, posing a serious threat to the survival of patients. Although the diagnosis and treatment of PSM have made significant progress in the past two decades, numerous challenges remain. Recently, functionalized biomaterials have shown promise for PSM diagnosis and treatment. Hence, we review the progress of functionalized biomaterials for the diagnosis and treatment of PSM. We first introduce the classification and pathogenesis of PSM. We then discuss the applications of functionalized biomaterials for the diagnosis and treatment of PSM. In particular, we focus on functionalized biomaterials as drug carriers for the treatment of PSM, including chemotherapy, immunotherapy, targeted therapy, combination therapy, and other therapies. Finally, we summarized the current challenges and provided a perspective on the diagnosis and treatment of PSM.
Collapse
Affiliation(s)
- Xiusen Qin
- Department of General SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Institute of GastroenterologyGuangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesBiomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Institute of Biomedical Innovation and Laboratory of Regenerative Medicine and BiomaterialsBiomedical Material Conversion and Evaluation Engineering Technology Research Center of Guangdong ProvinceGuangzhouChina
| | - Mingli Su
- Guangdong Institute of GastroenterologyGuangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesBiomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of Endoscopic SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Huili Guo
- Department of Infectious DiseasesThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Binying Peng
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Rui Luo
- Department of General SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Institute of GastroenterologyGuangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesBiomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Institute of Biomedical Innovation and Laboratory of Regenerative Medicine and BiomaterialsBiomedical Material Conversion and Evaluation Engineering Technology Research Center of Guangdong ProvinceGuangzhouChina
| | - Junwen Ye
- Department of General SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Institute of GastroenterologyGuangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesBiomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Institute of Biomedical Innovation and Laboratory of Regenerative Medicine and BiomaterialsBiomedical Material Conversion and Evaluation Engineering Technology Research Center of Guangdong ProvinceGuangzhouChina
| | - Hui Wang
- Department of General SurgeryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Institute of GastroenterologyGuangdong Provincial Key Laboratory of Colorectal and Pelvic Floor DiseasesBiomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Institute of Biomedical Innovation and Laboratory of Regenerative Medicine and BiomaterialsBiomedical Material Conversion and Evaluation Engineering Technology Research Center of Guangdong ProvinceGuangzhouChina
| |
Collapse
|
26
|
Wong LY, Lui NS. Intraoperative Molecular Imaging of Lung Cancer. Thorac Surg Clin 2023; 33:227-232. [PMID: 37414478 DOI: 10.1016/j.thorsurg.2023.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Intraoperative molecular imaging innovations have been propelled by the development of fluorescent contrast agents that specifically target tumor tissues and advanced camera systems that can detect the specified fluorescence. The most promising agent to date is OTL38, a targeted and near-infrared agent that was recently approved by the Food and Drug Administration for intraoperative imaging for lung cancer.
Collapse
Affiliation(s)
- Lye-Yeng Wong
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Falk Building, Stanford, CA 94305, USA. https://twitter.com/LyeYengWongMD
| | - Natalie S Lui
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Falk Building, Stanford, CA 94305, USA.
| |
Collapse
|
27
|
von Kroge PH, Duprée A. Fluorescent Imaging in Visceral Surgery: Current Opportunities and Future Perspectives. Visc Med 2023; 39:39-45. [PMID: 37405326 PMCID: PMC10315688 DOI: 10.1159/000530362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/21/2023] [Indexed: 07/06/2023] Open
Abstract
Background Fluorescent imaging using indocyanine green (FI-ICG) has become quite popular in the past century, giving the surgeon various pre- and intraoperative approaches in visceral surgery. Nevertheless, several aspects and pitfalls of using the technology need to be addressed. Summary This article focused on the applications of FI-ICG in esophageal and colorectal surgery as this is where the clinical relevance is most important. Important benchmark studies were summarized to explain the background. In addition, dosage, the timing of application, and future perspectives - especially quantification methods - were the article's content. Key Message There are currently encouraging data on the use of FI-ICG, particularly concerning perfusion assessment to reduce anastomotic leakage, although its use is mainly subjective. The optimal dosage remains unclear; for perfusion evaluation, it should be around 0.1 mg/kg body weight. Moreover, the quantification of FI-ICG opens new possibilities, so that reference values may be available in the future. However, in addition to perfusion measurement, the detection of additional hepatic lesions such as liver metastases or lesions of peritoneal carcinomatosis is also possible. A standardization of FI-ICG and further studies are needed to fully utilize FI-ICG.
Collapse
Affiliation(s)
- Philipp H von Kroge
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Duprée
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
28
|
Bortot B, Mangogna A, Di Lorenzo G, Stabile G, Ricci G, Biffi S. Image-guided cancer surgery: a narrative review on imaging modalities and emerging nanotechnology strategies. J Nanobiotechnology 2023; 21:155. [PMID: 37202750 DOI: 10.1186/s12951-023-01926-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023] Open
Abstract
Surgical resection is the cornerstone of solid tumour treatment. Current techniques for evaluating margin statuses, such as frozen section, imprint cytology, and intraoperative ultrasound, are helpful. However, an intraoperative assessment of tumour margins that is accurate and safe is clinically necessary. Positive surgical margins (PSM) have a well-documented negative effect on treatment outcomes and survival. As a result, surgical tumour imaging methods are now a practical method for reducing PSM rates and improving the efficiency of debulking surgery. Because of their unique characteristics, nanoparticles can function as contrast agents in image-guided surgery. While most image-guided surgical applications utilizing nanotechnology are now in the preclinical stage, some are beginning to reach the clinical phase. Here, we list the various imaging techniques used in image-guided surgery, such as optical imaging, ultrasound, computed tomography, magnetic resonance imaging, nuclear medicine imaging, and the most current developments in the potential of nanotechnology to detect surgical malignancies. In the coming years, we will see the evolution of nanoparticles tailored to specific tumour types and the introduction of surgical equipment to improve resection accuracy. Although the promise of nanotechnology for producing exogenous molecular contrast agents has been clearly demonstrated, much work remains to be done to put it into practice.
Collapse
Affiliation(s)
- Barbara Bortot
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Alessandro Mangogna
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giovanni Di Lorenzo
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Guglielmo Stabile
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Giuseppe Ricci
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Stefania Biffi
- Obstetrics and Gynecology, Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.
| |
Collapse
|
29
|
Sikkenk DJ, Sterkenburg AJ, Schmidt I, Gorpas D, Nagengast WB, Consten ECJ. Detection of Tumour-Targeted IRDye800CW Tracer with Commercially Available Laparoscopic Surgical Systems. Diagnostics (Basel) 2023; 13:diagnostics13091591. [PMID: 37174982 PMCID: PMC10178288 DOI: 10.3390/diagnostics13091591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
(1) Introduction: Near-infrared fluorescence (NIRF) combined with tumour-targeted tracers, such as bevacizumab-800CW, could aid surgical decision-making. This study explored the use of IRDye800CW, conjugated to bevacizumab, with four commercially available NIRF laparoscopes optimised for indocyanine green (ICG). (2) Methods: A (lymph node) phantom was made from a calibration device for NIRF and tissue-mimicking material. Serial dilutions of bevacizumab-800CW were made and ICG functioned as a reference. System settings, working distance, and thickness of tissue-mimicking material were varied to assess visibility of the fluorescence signal and tissue penetration. Tests were performed with four laparoscopes: VISERA ELITE II, Olympus; IMAGE1 S™ 4U Rubina, KARL STORZ; ENDOCAM Logic 4K platform, Richard Wolf; da Vinci Xi, Intuitive Surgical. (3) Results: The lowest visible bevacizumab-800CW concentration ranged between 13-850 nM (8-512 times diluted stock solution) for all laparoscopes, but the tracer was not visible through 0.8 cm of tissue in all systems. In contrast, ICG was still visible at a concentration of 0.4 nM (16,384 times diluted) and through 1.6-2.4 cm of tissue. Visibility and tissue penetration generally improved with a reduced working distance and manually adjusted system settings. (4) Conclusion: Depending on the application, bevacizumab-800CW might be sufficiently visible with current laparoscopes, but optimisation would widen applicability of tumour-targeted IRDye800CW tracers.
Collapse
Affiliation(s)
- Daan J Sikkenk
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Surgery, Meander Medical Centre, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| | - Andrea J Sterkenburg
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Dimitris Gorpas
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Ingolstädter Landstraße 1, D-85764 Neuherberg, Germany
- Chair of Biological Imaging, Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, Ismaninger Straße 22, D-81675 Munich, Germany
| | - Wouter B Nagengast
- Department of Gastroenterology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Esther C J Consten
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Surgery, Meander Medical Centre, Maatweg 3, 3813 TZ Amersfoort, The Netherlands
| |
Collapse
|
30
|
Mulder BGS, Koller M, Duiker EW, Sarasqueta AF, Burggraaf J, Meijer VED, Vahrmeijer AL, Hoogwater FJH, Bonsing BA, van Dam GM, Mieog JSD, Pranger BK. Intraoperative Molecular Fluorescence Imaging of Pancreatic Cancer by Targeting Vascular Endothelial Growth Factor: A Multicenter Feasibility Dose-Escalation Study. J Nucl Med 2023; 64:82-89. [PMID: 35680414 PMCID: PMC9841260 DOI: 10.2967/jnumed.121.263773] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 01/28/2023] Open
Abstract
Tumor visualization with near-infrared fluorescence (NIRF) imaging might aid exploration and resection of pancreatic cancer by visualizing the tumor in real time. Conjugation of the near-infrared fluorophore IRDye800CW to the monoclonal antibody bevacizumab enables targeting of vascular endothelial growth factor A. The aim of this study was to determine whether intraoperative tumor-specific imaging of pancreatic cancer with the fluorescent tracer bevacizumab-800CW is feasible and safe. Methods: In this multicenter dose-escalation phase I trial, patients in whom pancreatic ductal adenocarcinoma (PDAC) was suspected were administered bevacizumab-800CW (4.5, 10, or 25 mg) 3 d before surgery. Safety monitoring encompassed allergic or anaphylactic reactions and serious adverse events attributed to bevacizumab-800CW. Intraoperative NIRF imaging was performed immediately after laparotomy, just before and after resection of the specimen. Postoperatively, fluorescence signals on the axial slices and formalin-fixed paraffin-embedded tissue blocks from the resected specimens were correlated with histology. Subsequently, tumor-to-background ratios (TBR) were calculated. Results: Ten patients with clinically suspected PDAC were enrolled in the study. Four of the resected specimens were confirmed PDACs; other malignancies were distal cholangiocarcinoma, ampullary carcinoma, and neuroendocrine tumors. No serious adverse events were related to bevacizumab-800CW. In vivo tumor visualization with NIRF imaging differed per tumor type and was nonconclusive. Ex vivo TBRs were 1.3, 1.5, and 2.5 for the 4.5-, 10-, and 25-mg groups, respectively. Conclusion: NIRF-guided surgery in patients with suspected PDAC using bevacizumab-IRDye800CW is feasible and safe. However, suboptimal TBRs were obtained because no clear distinction between pancreatic cancer from normal or inflamed pancreatic tissue was achieved. Therefore, a more tumor-specific tracer than bevacizumab-IRDye800CW for PDAC is preferred.
Collapse
Affiliation(s)
| | - Marjory Koller
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Evelien W Duiker
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Vincent E de Meijer
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | | | - Frederik J H Hoogwater
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Gooitzen M van Dam
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- AxelaRx/TRACER Europe BV, Groningen, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Bobby K Pranger
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands;
| |
Collapse
|
31
|
Tian Z, Liang S, Zhou X, Luo H, Tian M, Zhang X, Guo C, Zhang J. Near-infrared-dye labeled tumor vascular-targeted dimer GEBP11 peptide for image-guided surgery in gastric cancer. Front Oncol 2022; 12:885036. [PMID: 36505820 PMCID: PMC9730820 DOI: 10.3389/fonc.2022.885036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Positive resection margins occur in about 2.8%-8.2% gastric cancer surgeries and is associated with poor prognosis. Intraoperative guidance using Nearinfrared (NIR) fluorescence imaging is a promising technique for tumor detection and margin assessment. The goal of this study was to develop a tumor-specific probe for real-time intraoperative NIR fluorescence imaging guidance. Methods The tumor vascular homing peptide specific for gastric cancer, GEBP11, was conjugated with a near-infrared fluorophore, Cy5.5. The binding specificity of the GEBP11 probes to tumor vascular endothelial cells were confirmed by immunofluorescent staining. The ability of the probe to detect tumor lesions was evaluated in two xenograft models. An orthotopic gastric cancer xenograft model was used to evaluate the efficacy of the GEBP11 NIR probes in real-time surgical guidance. Results In vitro assay suggested that both mono and dimeric GEBP11 NIR probes could bind specifically to tumor vascular epithelial cells, with dimeric peptides showed better affinity. In tumor xenograft mice, live imaging suggested that comparing with free Cy5.5 probe, significantly stronger NIR signals could be detected at the tumor site at 24-48h after injection of mono or dimeric GEBP11 probes. Dimeric GEBP11 probe showed prolonged and stronger NIR signals than mono GEBP11 probe. Biodistribution assay suggested that GEBP11 NIR probes were enriched in gastric cancer xenografts. Using dimeric GEBP11 NIR probes in real-time surgery, the tumor margins and peritoneal metastases could be clearly visualized. Histological examination confirmed the complete resection of the tumor. Conclusion (GEBP11)2-ACP-Cy5.5 could be a potential useful probe for intraoperative florescence guidance in gastric cancer surgery.
Collapse
Affiliation(s)
- Zuhong Tian
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China
| | - Shuhui Liang
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China
| | - Xinmin Zhou
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China
| | | | - Miaomiao Tian
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China
| | - Xianghan Zhang
- Engineering Research Center of Molecular-imaging and Neuroimaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, China
| | - Changcun Guo
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China,*Correspondence: Changcun Guo, ; Jing Zhang,
| | - Jing Zhang
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, China,*Correspondence: Changcun Guo, ; Jing Zhang,
| |
Collapse
|
32
|
Refaat A, Yap ML, Pietersz G, Walsh APG, Zeller J, Del Rosal B, Wang X, Peter K. In vivo fluorescence imaging: success in preclinical imaging paves the way for clinical applications. J Nanobiotechnology 2022; 20:450. [PMID: 36243718 PMCID: PMC9571426 DOI: 10.1186/s12951-022-01648-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
Advances in diagnostic imaging have provided unprecedented opportunities to detect diseases at early stages and with high reliability. Diagnostic imaging is also crucial to monitoring the progress or remission of disease and thus is often the central basis of therapeutic decision-making. Currently, several diagnostic imaging modalities (computed tomography, magnetic resonance imaging, and positron emission tomography, among others) are routinely used in clinics and present their own advantages and limitations. In vivo near-infrared (NIR) fluorescence imaging has recently emerged as an attractive imaging modality combining low cost, high sensitivity, and relative safety. As a preclinical tool, it can be used to investigate disease mechanisms and for testing novel diagnostics and therapeutics prior to their clinical use. However, the limited depth of tissue penetration is a major challenge to efficient clinical use. Therefore, the current clinical use of fluorescence imaging is limited to a few applications such as image-guided surgery on tumors and retinal angiography, using FDA-approved dyes. Progress in fluorophore development and NIR imaging technologies holds promise to extend their clinical application to oncology, cardiovascular diseases, plastic surgery, and brain imaging, among others. Nanotechnology is expected to revolutionize diagnostic in vivo fluorescence imaging through targeted delivery of NIR fluorescent probes using antibody conjugation. In this review, we discuss the latest advances in in vivo fluorescence imaging technologies, NIR fluorescent probes, and current and future clinical applications.
Collapse
Affiliation(s)
- Ahmed Refaat
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Engineering Technologies, Swinburne University of Technology, Melbourne, VIC, Australia.,Pharmaceutics Department, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - May Lin Yap
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geoffrey Pietersz
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Burnet Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Aidan Patrick Garing Walsh
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Johannes Zeller
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Plastic and Hand Surgery, University of Freiburg Medical Center, Freiburg, Germany
| | | | - Xiaowei Wang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia. .,Department of Medicine, Monash University, Melbourne, VIC, Australia. .,Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia.
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia. .,Department of Medicine, Monash University, Melbourne, VIC, Australia. .,Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
33
|
Detection of Experimental Colorectal Peritoneal Metastases by a Novel PDGFRβ-Targeting Nanobody. Cancers (Basel) 2022; 14:cancers14184348. [PMID: 36139509 PMCID: PMC9497196 DOI: 10.3390/cancers14184348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Colorectal cancer can metastasize to multiple distant sites. Metastases growing within the peritoneal cavity cause a high degree of morbidity and are associated with very poor survival. Moreover, peritoneal metastases are difficult to detect using conventional imaging methods. Consequently, peritoneal metastases are generally under-diagnosed and their response to therapy is difficult to assess. An extensive molecular and cellular analysis of colorectal peritoneal metastases revealed that these lesions express very high levels of specific markers that could serve as targets for imaging-based diagnosis and treatment. In the present report, we explore the potential value of one such marker, PDGFRB, to serve as a target for peritoneal metastasis detection by molecular imaging. Therefore, we generated a PDGFRB-binding llama nanobody and demonstrate its utility in detecting peritoneal metastases in mice. The clinical development of PDGFRB-targeting tracers may help to improve the diagnosis of peritoneal metastases and the clinical management of this highly aggressive disease entity. Abstract Peritoneal metastases in colorectal cancer (CRC) belong to Consensus Molecular Subtype 4 (CMS4) and are associated with poor prognosis. Conventional imaging modalities, such as Computed Tomography (CT) and Fluorodeoxyglucose-Positron Emission Tomography (FDG-PET), perform very poorly in the detection of peritoneal metastases. However, the stroma-rich nature of these lesions provides a basis for developing molecular imaging strategies. In this study, conducted from 2019 to 2021, we aimed to generate a Platelet-Derived Growth Factor Receptor beta (PDGFRB)-binding molecular imaging tracer for the detection of CMS4 CRC, including peritoneal metastases. The expression of PDGFRB mRNA discriminated CMS4 from CMS1-3 (AUROC = 0.86 (95% CI 0.85–0.88)) and was associated with poor relapse-free survival. PDGFRB mRNA and protein levels were very high in all human peritoneal metastases examined (n = 66). Therefore, we generated a PDGFRB-targeting llama nanobody (VHH1E12). Biotin-labelled VHH1E12 bound to immobilized human and mouse PDGFRB with high affinity (EC50 human PDGFRB = 7 nM; EC50 murine PDGFRB = 0.8 nM), and to PDGFRB-expressing HEK293 cells grown in vitro. A pharmacokinetic analysis of IRDye-800CW-conjugated VHH1E12 in mice showed that the plasma half-life was 6 min. IRDye-800CW-conjugated VHH1E12 specifically accumulated in experimentally induced colorectal cancer peritoneal metastases in mice. A tissue analysis subsequently demonstrated co-localization of the nanobody with PDGFRB expression in the tumour stroma. Our results demonstrate the potential value of PDGFRB-targeted molecular imaging as a novel strategy for the non-invasive detection of CMS4 CRC, in particular, peritoneal metastases.
Collapse
|
34
|
Choi JH, Kang CM, Park JY. EGFR-targeted fluorescent imaging using the da Vinci® Firefly™ camera for gallbladder cancer. World J Surg Oncol 2022; 20:201. [PMID: 35701793 PMCID: PMC9199159 DOI: 10.1186/s12957-022-02675-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/05/2022] [Indexed: 11/29/2022] Open
Abstract
Background Fluorescent imaging may aid with the precise diagnosis and treatment of patients with gallbladder cancer. In this study, we sought to demonstrate whether the da Vinci® surgical system and Firefly™ camera could detect EGFR-targeted fluorescent images in orthotopic mouse models of gallbladder cancer. Methods An orthotopic mouse model of gallbladder cancer was created by injecting NOZ gallbladder cancer cells mixed with Matrigel into the gallbladder. In vivo imaging of subcutaneous and orthotopic gallbladder tumors was performed after the injection of DyLight 650- or 800-conjugated EGFR antibody. Results Western blotting, flow cytometry, and confocal microscopy showed the presence of EGFR in NOZ cells, but not in HEK293 cells. Subcutaneous NOZ cell tumors fluoresced after injection with fluorescent EGFR antibody, but subcutaneous HEK293 tumors did not. Fluorescent EGFR antibody made orthotopic NOZ tumors fluoresce, with an intensity stronger than that in the surrounding normal tissues. Histochemical examination confirmed the location of the tumors inside the gallbladder and adjacent liver parenchyma. Fluorescent signal was also detected in orthotopic gallbladder tumors with Firefly™ camera. Conclusion Our study showed that fluorescent EGFR antibodies and the Firefly camera in the da Vinci system can detect fluorescing gallbladder tumors, which demonstrates their potential use for molecular imaging-based prevision surgery in the near future.
Collapse
Affiliation(s)
- Jung Ha Choi
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Chang Moo Kang
- Division of HBP Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Pancreatobiliary Cancer Center, Yonsei Cancer Center, and Yonsei Institute of Gastroenterology, Severance Hospital, Seoul, South Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea. .,Pancreatobiliary Cancer Center, Yonsei Cancer Center, and Yonsei Institute of Gastroenterology, Severance Hospital, Seoul, South Korea.
| |
Collapse
|
35
|
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: 3.3] [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
|
36
|
Vergeer RA, Theunissen REP, van Elk T, Schmidt I, Postma MR, Tamasi K, van Dijk JMC, Kuijlen JMA. Fluorescence-guided detection of pituitary neuroendocrine tumor (PitNET) tissue during endoscopic transsphenoidal surgery available agents, their potential, and technical aspects. Rev Endocr Metab Disord 2022; 23:647-657. [PMID: 35344185 PMCID: PMC9156450 DOI: 10.1007/s11154-022-09718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2022] [Indexed: 10/29/2022]
Abstract
Differentiation of pituitary neuroendocrine tumor (PitNET) tissue from surrounding normal tissue during surgery is challenging. A number of fluorescent agents is available for visualization of tissue discrepancy, with the potential of improving total tumor resection. This review evaluates the availability, clinical and technical applicability of the various fluorescent agents within the field of pituitary surgery. According to PRISMA guidelines, a systematic review was performed to identify reports describing results of in vivo application of fluorescent agents. In this review, 15 publications were included. Sodium Fluorescein (FNa) was considered in two studies. The first study reported noticeable fluorescence in adenoma tissue, the second demonstrated the strongest fluorescence in non-functioning pituitary adenomas. 5-Aminolevulinic acid (5-ALA) was investigated in three studies. One study compared laser-based optical biopsy system (OBS) with photo-diagnostic filter (PD) and found that the OBS was able to detect all microadenomas, even when MRI was negative. The second study retrospectively analyzed twelve pituitary adenomas and found only one positive for fluorescence. The third investigated fifteen pituitary adenomas of which one displayed vague fluorescence. Indocyanine green (ICG) was researched in four studies with variable results. Second-Window ICG yielded no significant difference between functioning and non-functioning adenomas in one study, while a second study displayed 4 times higher fluorescence in tumor tissue than in normal tissue. In three studies, OTL38 showed potential in non-functioning pituitary adenomas. At present, evidence for fluorescent agents to benefit total resection of PitNETs is lacking. OTL38 can potentially serve as a selective fluorescent agent in non-functioning pituitary adenomas in the near future.
Collapse
Affiliation(s)
- Rob A Vergeer
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Robin E P Theunissen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Theodora van Elk
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iris Schmidt
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mark R Postma
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Katalin Tamasi
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J Marc C van Dijk
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jos M A Kuijlen
- Department of Neurosurgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
37
|
Multimodal CEA-targeted fluorescence and radioguided cytoreductive surgery for peritoneal metastases of colorectal origin. Nat Commun 2022; 13:2621. [PMID: 35551444 PMCID: PMC9098887 DOI: 10.1038/s41467-022-29630-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 03/16/2022] [Indexed: 11/09/2022] Open
Abstract
In patients with colorectal peritoneal metastases scheduled for cytoreductive surgery, accurate preoperative estimation of tumor burden and subsequent intraoperative detection of all tumor deposits remains challenging. In this study (ClinicalTrials.gov NCT03699332) we describe the results of a phase I clinical trial evaluating [111In]In-DOTA-labetuzumab-IRDye800CW, a dual-labeled anti-carcinoembryonic antigen (anti-CEA) antibody conjugate that enables both preoperative imaging and intraoperative radioguidance and fluorescence imaging. Primary study outcomes are safety and feasibility of this multimodal imaging approach. Secondary outcomes are determination of the optimal dose, correlation between tracer uptake and histopathology and effects on clinical strategy. Administration of [111In]In-DOTA-labetuzumab-IRDye800CW is well-tolerated and enables sensitive pre- and intraoperative imaging in patients who receive 10 or 50 mg of the tracer. Preoperative imaging revealed previously undetected lymph node metastases in one patient, and intraoperative fluorescence imaging revealed four previously undetected metastases in two patients. Alteration of clinical strategy based on multimodal imaging occurred in three patients. Thus, multimodal image-guided surgery after administration of this dual-labeled tracer is a promising approach that may aid in decision making before and during cytoreductive surgical procedures.
Collapse
|
38
|
Josserand V, Bernard C, Michy T, Guidetti M, Vollaire J, Coll JL, Hurbin A. Tumor-Specific Imaging with Angiostamp800 or Bevacizumab-IRDye 800CW Improves Fluorescence-Guided Surgery over Indocyanine Green in Peritoneal Carcinomatosis. Biomedicines 2022; 10:biomedicines10051059. [PMID: 35625796 PMCID: PMC9138305 DOI: 10.3390/biomedicines10051059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/16/2022] Open
Abstract
Complete surgical removal of lesions improves survival of peritoneal carcinomatosis and can be enhanced by intraoperative near-infrared fluorescence imaging. Indocyanine green (ICG) is the only near-infrared fluorescent dye approved for clinical use, but it lacks specificity for tumor cells, highlighting the need for tumor-selective targeting agents. We compared the tumor-specific near-infrared fluorescent probes Bevacizumab-IRDye 800CW and Angiostamp800, which target tumor angiogenesis and cancer cells, to ICG for fluorescence-guided surgery in peritoneal carcinomatosis of ovarian origin. The probes were administered to mice with orthotopic peritoneal carcinomatosis prior to conventional and fluorescence-guided surgery. The influence of neoadjuvant chemotherapy was also assessed. Conventional surgery removed 88.0 ± 1.2% of the total tumor load in mice. Fluorescence-guided surgery allowed the resection of additional nodules, enhancing the total tumor burden resection by 9.8 ± 0.7%, 8.5 ± 0.8%, and 3.9 ± 1.2% with Angiostamp800, Bevacizumab-IRDye 800CW and ICG, respectively. Interestingly, among the resected nodules, 15% were false-positive with ICG, compared to only 1.4% with Angiostamp800 and 3.5% with Bevacizumab-IRDye 800CW. Furthermore, conventional surgery removed only 69.0 ± 3.9% of the total tumor burden after neoadjuvant chemotherapy. Fluorescence-guided surgery with Angiostamp800 and Bevacizumab-IRDye 800CW increased the total tumor burden resection to 88.7 ± 4.3%, whereas ICG did not improve surgery at all. Bevacizumab-IRDye 800CW and Angiostamp800 better detect ovarian tumors and metastases than the clinically used fluorescent tracer ICG, and can help surgeons completely remove tumors, especially after surgery neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Véronique Josserand
- Institute for Advanced Biosciences, Institut National de la Santé et de la Recherche Médicale INSERM U1209, Centre National de la Recherche Scientifique CNRS UMR5309, Université Grenoble Alpes, F-38000 Grenoble, France; (V.J.); (C.B.); (T.M.); (M.G.); (J.V.); (J.-L.C.)
| | - Claire Bernard
- Institute for Advanced Biosciences, Institut National de la Santé et de la Recherche Médicale INSERM U1209, Centre National de la Recherche Scientifique CNRS UMR5309, Université Grenoble Alpes, F-38000 Grenoble, France; (V.J.); (C.B.); (T.M.); (M.G.); (J.V.); (J.-L.C.)
- Centre Hospitalier Universitaire Grenoble Alpes, Université Grenoble Alpes, F-38000 Grenoble, France
| | - Thierry Michy
- Institute for Advanced Biosciences, Institut National de la Santé et de la Recherche Médicale INSERM U1209, Centre National de la Recherche Scientifique CNRS UMR5309, Université Grenoble Alpes, F-38000 Grenoble, France; (V.J.); (C.B.); (T.M.); (M.G.); (J.V.); (J.-L.C.)
- Centre Hospitalier Universitaire Grenoble Alpes, Université Grenoble Alpes, F-38000 Grenoble, France
| | - Mélanie Guidetti
- Institute for Advanced Biosciences, Institut National de la Santé et de la Recherche Médicale INSERM U1209, Centre National de la Recherche Scientifique CNRS UMR5309, Université Grenoble Alpes, F-38000 Grenoble, France; (V.J.); (C.B.); (T.M.); (M.G.); (J.V.); (J.-L.C.)
| | - Julien Vollaire
- Institute for Advanced Biosciences, Institut National de la Santé et de la Recherche Médicale INSERM U1209, Centre National de la Recherche Scientifique CNRS UMR5309, Université Grenoble Alpes, F-38000 Grenoble, France; (V.J.); (C.B.); (T.M.); (M.G.); (J.V.); (J.-L.C.)
| | - Jean-Luc Coll
- Institute for Advanced Biosciences, Institut National de la Santé et de la Recherche Médicale INSERM U1209, Centre National de la Recherche Scientifique CNRS UMR5309, Université Grenoble Alpes, F-38000 Grenoble, France; (V.J.); (C.B.); (T.M.); (M.G.); (J.V.); (J.-L.C.)
| | - Amandine Hurbin
- Institute for Advanced Biosciences, Institut National de la Santé et de la Recherche Médicale INSERM U1209, Centre National de la Recherche Scientifique CNRS UMR5309, Université Grenoble Alpes, F-38000 Grenoble, France; (V.J.); (C.B.); (T.M.); (M.G.); (J.V.); (J.-L.C.)
- Correspondence:
| |
Collapse
|
39
|
Voskuil FJ, Vonk J, van der Vegt B, Kruijff S, Ntziachristos V, van der Zaag PJ, Witjes MJH, van Dam GM. Intraoperative imaging in pathology-assisted surgery. Nat Biomed Eng 2022; 6:503-514. [PMID: 34750537 DOI: 10.1038/s41551-021-00808-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
The pathological assessment of surgical specimens during surgery can reduce the incidence of positive resection margins, which otherwise can result in additional surgeries or aggressive therapeutic regimens. To improve patient outcomes, intraoperative spectroscopic, fluorescence-based, structural, optoacoustic and radiological imaging techniques are being tested on freshly excised tissue. The specific clinical setting and tumour type largely determine whether endogenous or exogenous contrast is to be detected and whether the tumour specificity of the detected biomarker, image resolution, image-acquisition times or penetration depth are to be prioritized. In this Perspective, we describe current clinical standards for intraoperative tissue analysis and discuss how intraoperative imaging is being implemented. We also discuss potential implementations of intraoperative pathology-assisted surgery for clinical decision-making.
Collapse
Affiliation(s)
- Floris J Voskuil
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jasper Vonk
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Schelto Kruijff
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vasilis Ntziachristos
- Chair for Biological Imaging, Center for Translational Cancer Research, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
| | - Pieter J van der Zaag
- Phillips Research Laboratories, Eindhoven, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Molecular Biophysics, Zernike Institute, University of Groningen, Groningen, The Netherlands
| | - Max J H Witjes
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gooitzen M van Dam
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. .,AxelaRx/TRACER BV, Groningen, The Netherlands.
| |
Collapse
|
40
|
Wang F, Qu L, Ren F, Baghdasaryan A, Jiang Y, Hsu R, Liang P, Li J, Zhu G, Ma Z, Dai H. High-precision tumor resection down to few-cell level guided by NIR-IIb molecular fluorescence imaging. Proc Natl Acad Sci U S A 2022; 119:e2123111119. [PMID: 35380898 PMCID: PMC9169804 DOI: 10.1073/pnas.2123111119] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/03/2022] [Indexed: 12/19/2022] Open
Abstract
In vivo fluorescence/luminescence imaging in the near-infrared-IIb (NIR-IIb, 1,500 to 1,700 nm) window under <1,000 nm excitation can afford subcentimeter imaging depth without any tissue autofluorescence, promising high-precision intraoperative navigation in the clinic. Here, we developed a compact imager for concurrent visible photographic and NIR-II (1,000 to 3,000 nm) fluorescence imaging for preclinical image-guided surgery. Biocompatible erbium-based rare-earth nanoparticles (ErNPs) with bright down-conversion luminescence in the NIR-IIb window were conjugated to TRC105 antibody for molecular imaging of CD105 angiogenesis markers in 4T1 murine breast tumors. Under a ∼940 ± 38 nm light-emitting diode (LED) excitation, NIR-IIb imaging of 1,500- to 1,700-nm emission afforded noninvasive tumor–to–normal tissue (T/NT) signal ratios of ∼40 before surgery and an ultrahigh intraoperative tumor-to-muscle (T/M) ratio of ∼300, resolving tumor margin unambiguously without interfering background signal from surrounding healthy tissues. High-resolution imaging resolved small numbers of residual cancer cells during surgery, allowing thorough and nonexcessive tumor removal at the few-cell level. NIR-IIb molecular imaging afforded 10-times-higher and 100-times-higher T/NT and T/M ratios, respectively, than imaging with IRDye800CW-TRC105 in the ∼900- to 1,300-nm range. The vastly improved resolution of tumor margin and diminished background open a paradigm of molecular imaging-guided surgery.
Collapse
Affiliation(s)
- Feifei Wang
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - Liangqiong Qu
- School of Medicine, Stanford University, Stanford, CA 94303
| | - Fuqiang Ren
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - Ani Baghdasaryan
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - Yingying Jiang
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - RuSiou Hsu
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - Peng Liang
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - Jiachen Li
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - Guanzhou Zhu
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - Zhuoran Ma
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| | - Hongjie Dai
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Bio-X, Stanford University, Stanford, CA 94305
| |
Collapse
|
41
|
Yang E, Liu Q, Huang G, Liu J, Wei W. Engineering nanobodies for next-generation molecular imaging. Drug Discov Today 2022; 27:1622-1638. [PMID: 35331925 DOI: 10.1016/j.drudis.2022.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/04/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
In recent years, nanobodies have emerged as ideal imaging agents for molecular imaging. Molecular nanobody imaging combines the specificity of nanobodies with the sensitivity of state-of-the-art molecular imaging modalities, such as positron emission tomography (PET). Given that modifications of nanobodies alter their pharmacokinetics (PK), the engineering strategies that combine nanobodies with radionuclides determine the effectiveness, reliability, and safety of the molecular imaging probes. In this review, we introduce conjugation strategies that have been applied to nanobodies, including random conjugation, 99mTc tricarbonyl chemistry, sortase A-mediated site-specific conjugation, maleimide-cysteine chemistry, and click chemistries. We also summarize the latest advances in nanobody tracers, emphasizing their preclinical and clinical use. In addition, we elaborate on nanobody-based near-infrared fluorescence (NIRF) imaging and image-guided surgery.
Collapse
Affiliation(s)
- Erpeng Yang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| |
Collapse
|
42
|
Fluorescence Molecular Targeting of Colon Cancer to Visualize the Invisible. Cells 2022; 11:cells11020249. [PMID: 35053365 PMCID: PMC8773892 DOI: 10.3390/cells11020249] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/28/2021] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) is a common cause of cancer and cancer-related death. Surgery is the only curative modality. Fluorescence-enhanced visualization of CRC with targeted fluorescent probes that can delineate boundaries and target tumor-specific biomarkers can increase rates of curative resection. Approaches to enhancing visualization of the tumor-to-normal tissue interface are active areas of investigation. Nonspecific dyes are the most-used approach, but tumor-specific targeting agents are progressing in clinical trials. The present narrative review describes the principles of fluorescence targeting of CRC for diagnosis and fluorescence-guided surgery with molecular biomarkers for preclinical or clinical evaluation.
Collapse
|
43
|
Fundamentals and developments in fluorescence-guided cancer surgery. Nat Rev Clin Oncol 2022; 19:9-22. [PMID: 34493858 DOI: 10.1038/s41571-021-00548-3] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2021] [Indexed: 02/07/2023]
Abstract
Fluorescence-guided surgery using tumour-targeted imaging agents has emerged over the past decade as a promising and effective method of intraoperative cancer detection. An impressive number of fluorescently labelled antibodies, peptides, particles and other molecules related to cancer hallmarks have been developed for the illumination of target lesions. New approaches are being implemented to translate these imaging agents into the clinic, although only a few have made it past early-phase clinical trials. For this translational process to succeed, target selection, imaging agents and their related detection systems and clinical implementation have to operate in perfect harmony to enable real-time intraoperative visualization that can benefit patients. Herein, we review key aspects of this imaging cascade and focus on imaging approaches and methods that have helped to shed new light onto the field of intraoperative fluorescence-guided cancer surgery with the singular goal of improving patient outcomes.
Collapse
|
44
|
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
|
45
|
Kim YJ, Kim CH. Treatment for Peritoneal Metastasis of Patients With Colorectal Cancer. Ann Coloproctol 2021; 37:425-433. [PMID: 34961304 PMCID: PMC8717073 DOI: 10.3393/ac.2021.00920.0131] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
From the perspective of survival outcomes, the cancer survival of colorectal cancer (CRC) in the whole stage has improved. Peritoneal metastasis (PM) is found in approximately 8% to 15% of patients with CRC, with a poorer prognosis than that associated with other sites of metastases. Randomized controlled trials and up-to-date meta-analyses provide firm evidence that cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) could significantly improve overall survival compared with systemic chemotherapy alone in selected patients with CRC-PM. Practical guidelines recommend that the management of CRC-PM should be led by a multidisciplinary team carried out in experienced centers and consider CRS plus HIPEC for selected patients. In this review, we aim to provide the latest results of land mark studies and an overview of recent insights with regard to the management of CRC-PM.
Collapse
Affiliation(s)
- Young Jin Kim
- Department of Surgery, Seokjeong Wellpark Hospital, Gochang, Korea
| | - Chang Hyun Kim
- Division of Colorectal Surgery, Department of Surgery, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
46
|
van Dam MA, Vuijk FA, Stibbe JA, Houvast RD, Luelmo SAC, Crobach S, Shahbazi Feshtali S, de Geus-Oei LF, Bonsing BA, Sier CFM, Kuppen PJK, Swijnenburg RJ, Windhorst AD, Burggraaf J, Vahrmeijer AL, Mieog JSD. Overview and Future Perspectives on Tumor-Targeted Positron Emission Tomography and Fluorescence Imaging of Pancreatic Cancer in the Era of Neoadjuvant Therapy. Cancers (Basel) 2021; 13:6088. [PMID: 34885196 PMCID: PMC8656821 DOI: 10.3390/cancers13236088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Despite recent advances in the multimodal treatment of pancreatic ductal adenocarcinoma (PDAC), overall survival remains poor with a 5-year cumulative survival of approximately 10%. Neoadjuvant (chemo- and/or radio-) therapy is increasingly incorporated in treatment strategies for patients with (borderline) resectable and locally advanced disease. Neoadjuvant therapy aims to improve radical resection rates by reducing tumor mass and (partial) encasement of important vascular structures, as well as eradicating occult micrometastases. Results from recent multicenter clinical trials evaluating this approach demonstrate prolonged survival and increased complete surgical resection rates (R0). Currently, tumor response to neoadjuvant therapy is monitored using computed tomography (CT) following the RECIST 1.1 criteria. Accurate assessment of neoadjuvant treatment response and tumor resectability is considered a major challenge, as current conventional imaging modalities provide limited accuracy and specificity for discrimination between necrosis, fibrosis, and remaining vital tumor tissue. As a consequence, resections with tumor-positive margins and subsequent early locoregional tumor recurrences are observed in a substantial number of patients following surgical resection with curative intent. Of these patients, up to 80% are diagnosed with recurrent disease after a median disease-free interval of merely 8 months. These numbers underline the urgent need to improve imaging modalities for more accurate assessment of therapy response and subsequent re-staging of disease, thereby aiming to optimize individual patient's treatment strategy. In cases of curative intent resection, additional intra-operative real-time guidance could aid surgeons during complex procedures and potentially reduce the rate of incomplete resections and early (locoregional) tumor recurrences. In recent years intraoperative imaging in cancer has made a shift towards tumor-specific molecular targeting. Several important molecular targets have been identified that show overexpression in PDAC, for example: CA19.9, CEA, EGFR, VEGFR/VEGF-A, uPA/uPAR, and various integrins. Tumor-targeted PET/CT combined with intraoperative fluorescence imaging, could provide valuable information for tumor detection and staging, therapy response evaluation with re-staging of disease and intraoperative guidance during surgical resection of PDAC. METHODS A literature search in the PubMed database and (inter)national trial registers was conducted, focusing on studies published over the last 15 years. Data and information of eligible articles regarding PET/CT as well as fluorescence imaging in PDAC were reviewed. Areas covered: This review covers the current strategies, obstacles, challenges, and developments in targeted tumor imaging, focusing on the feasibility and value of PET/CT and fluorescence imaging for integration in the work-up and treatment of PDAC. An overview is given of identified targets and their characteristics, as well as the available literature of conducted and ongoing clinical and preclinical trials evaluating PDAC-targeted nuclear and fluorescent tracers.
Collapse
Affiliation(s)
- Martijn A. van Dam
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Floris A. Vuijk
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Judith A. Stibbe
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Ruben D. Houvast
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Saskia A. C. Luelmo
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Stijn Crobach
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | | | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, University Medical Center Leiden, 2333 ZA Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | | | - Albert D. Windhorst
- Department of Radiology, Section of Nuclear Medicine, Amsterdam UMC, Location VUmc, 1081 HV Amsterdam, The Netherlands;
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
- Centre for Human Drug Research, 2333 CL Leiden, The Netherlands
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| | - J. Sven D. Mieog
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.V.); (J.A.S.); (R.D.H.); (B.A.B.); (C.F.M.S.); (P.J.K.K.); (J.B.); (A.L.V.); (J.S.D.M.)
| |
Collapse
|
47
|
Turner MA, Lwin TM, Amirfakhri S, Nishino H, Hoffman RM, Yazaki PJ, Bouvet M. The Use of Fluorescent Anti-CEA Antibodies to Label, Resect and Treat Cancers: A Review. Biomolecules 2021; 11:1819. [PMID: 34944463 PMCID: PMC8699160 DOI: 10.3390/biom11121819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/21/2021] [Accepted: 11/26/2021] [Indexed: 02/07/2023] Open
Abstract
A major barrier to the diagnosis and effective treatment of solid-tumor cancers is the difficulty in detection and visualization of tumor margins in primary and metastatic disease. The use of fluorescence can augment the surgeon's ability to detect cancer and aid in its resection. Several cancer types express carcinoembryonic antigen (CEA) including colorectal, pancreatic and gastric cancer. Antibodies to CEA have been developed and tagged with near-infrared fluorescent dyes. This review article surveyed the use of CEA antibodies conjugated to fluorescent probes for in vivo studies since 1990. PubMed and Google Scholar databases were queried, and 900 titles and abstracts were screened. Fifty-nine entries were identified as possibly meeting inclusion/exclusion criteria and were reviewed in full. Forty articles were included in the review and their citations were screened for additional entries. A total of 44 articles were included in the final review. The use of fluorescent anti-CEA antibodies has been shown to improve detection and resection of tumors in both murine models and clinically. The cumulative results indicate that fluorescent-conjugated anti-CEA antibodies have important potential to improve cancer diagnosis and surgery. In an emerging technology, anti-CEA fluorescent antibodies have also been successfully used for photoimmunotherapy treatment for cancer.
Collapse
Affiliation(s)
- Michael A. Turner
- VA San Diego Healthcare System, La Jolla, CA 92161, USA; (M.A.T.); (S.A.); (H.N.); (R.M.H.)
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA
| | | | - Siamak Amirfakhri
- VA San Diego Healthcare System, La Jolla, CA 92161, USA; (M.A.T.); (S.A.); (H.N.); (R.M.H.)
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA
| | - Hiroto Nishino
- VA San Diego Healthcare System, La Jolla, CA 92161, USA; (M.A.T.); (S.A.); (H.N.); (R.M.H.)
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA
| | - Robert M. Hoffman
- VA San Diego Healthcare System, La Jolla, CA 92161, USA; (M.A.T.); (S.A.); (H.N.); (R.M.H.)
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA
- AntiCancer Inc., San Diego, CA 92111, USA
| | - Paul J. Yazaki
- Department of Immunology and Theranostics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA;
| | - Michael Bouvet
- VA San Diego Healthcare System, La Jolla, CA 92161, USA; (M.A.T.); (S.A.); (H.N.); (R.M.H.)
- Division of Surgical Oncology, Department of Surgery, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
48
|
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: 5.8] [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
|
49
|
Galema HA, Meijer RPJ, Lauwerends LJ, Verhoef C, Burggraaf J, Vahrmeijer AL, Hutteman M, Keereweer S, Hilling DE. Fluorescence-guided surgery in colorectal cancer; A review on clinical results and future perspectives. Eur J Surg Oncol 2021; 48:810-821. [PMID: 34657780 DOI: 10.1016/j.ejso.2021.10.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Colorectal cancer is the fourth most diagnosed malignancy worldwide and surgery is one of the cornerstones of the treatment strategy. Near-infrared (NIR) fluorescence imaging is a new and upcoming technique, which uses an NIR fluorescent agent combined with a specialised camera that can detect light in the NIR range. It aims for more precise surgery with improved oncological outcomes and a reduction in complications by improving discrimination between different structures. METHODS A systematic search was conducted in the Embase, Medline and Cochrane databases with search terms corresponding to 'fluorescence-guided surgery', 'colorectal surgery', and 'colorectal cancer' to identify all relevant trials. RESULTS The following clinical applications of fluorescence guided surgery for colorectal cancer were identified and discussed: (1) tumour imaging, (2) sentinel lymph node imaging, (3) imaging of distant metastases, (4) imaging of vital structures, (5) imaging of perfusion. Both experimental and FDA/EMA approved fluorescent agents are debated. Furthermore, promising future modalities are discussed. CONCLUSION Fluorescence-guided surgery for colorectal cancer is a rapidly evolving field. The first studies show additional value of this technique regarding change in surgical management. Future trials should focus on patient related outcomes such as complication rates, disease free survival, and overall survival.
Collapse
Affiliation(s)
- Hidde A Galema
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands; Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands
| | - Ruben P J Meijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands; Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, the Netherlands
| | - Lorraine J Lauwerends
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands; Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, the Netherlands
| | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands
| | - Merlijn Hutteman
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands
| | - Stijn Keereweer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands
| | - Denise E Hilling
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands; Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, the Netherlands.
| |
Collapse
|
50
|
Rijs Z, Jeremiasse B, Shifai N, Gelderblom H, Sier CFM, Vahrmeijer AL, van Leeuwen FWB, van der Steeg AFW, van de Sande MAJ. Introducing Fluorescence-Guided Surgery for Pediatric Ewing, Osteo-, and Rhabdomyosarcomas: A Literature Review. Biomedicines 2021; 9:biomedicines9101388. [PMID: 34680505 PMCID: PMC8533294 DOI: 10.3390/biomedicines9101388] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023] Open
Abstract
Sarcomas are a rare heterogeneous group of malignant neoplasms of mesenchymal origin which represent approximately 13% of all cancers in pediatric patients. The most prevalent pediatric bone sarcomas are osteosarcoma (OS) and Ewing sarcoma (ES). Rhabdomyosarcoma (RMS) is the most frequently occurring pediatric soft tissue sarcoma. The median age of OS and ES is approximately 17 years, so this disease is also commonly seen in adults while non-pleiomorphic RMS is rare in the adult population. The mainstay of all treatment regimens is multimodal treatment containing chemotherapy, surgical resection, and sometimes (neo)adjuvant radiotherapy. A clear resection margin improves both local control and overall survival and should be the goal during surgery with a curative intent. Real-time intraoperative fluorescence-guided imaging could facilitate complete resections by visualizing tumor tissue during surgery. This review evaluates whether non-targeted and targeted fluorescence-guided surgery (FGS) could be beneficial for pediatric OS, ES, and RMS patients. Necessities for clinical implementation, current literature, and the positive as well as negative aspects of non-targeted FGS using the NIR dye Indocyanine Green (ICG) were evaluated. In addition, we provide an overview of targets that could potentially be used for FGS in OS, ES, and RMS. Then, due to the time- and cost-efficient translational perspective, we elaborate on the use of antibody-based tracers as well as their disadvantages and alternatives. Finally, we conclude with recommendations for the experiments needed before FGS can be implemented for pediatric OS, ES, and RMS patients.
Collapse
Affiliation(s)
- Zeger Rijs
- Department of Orthopedic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.S.); (M.A.J.v.d.S.)
- Correspondence: ; Tel.: +31-641-637-074
| | - Bernadette Jeremiasse
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (B.J.); (A.F.W.v.d.S.)
| | - Naweed Shifai
- Department of Orthopedic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.S.); (M.A.J.v.d.S.)
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands;
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (C.F.M.S.); (A.L.V.)
- Percuros BV, 2333 CL Leiden, The Netherlands
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (C.F.M.S.); (A.L.V.)
| | - Fijs W. B. van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands;
| | - Alida F. W. van der Steeg
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (B.J.); (A.F.W.v.d.S.)
| | - Michiel A. J. van de Sande
- Department of Orthopedic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.S.); (M.A.J.v.d.S.)
| |
Collapse
|