1
|
Marrone S, Scalia G, Atallah O, Scibilia G, Palmisciano P, Chaurasia B, Maugeri R, Iacopino DG, Graziano F, Mastronardi L, Tomasi OS, Naimo J, Ferini G, Scollo P, Umana GE. Exploit the γ-Glutamyl hydroxymethyl rhodamine green fluorescence in surgical resection: A systematic literature review on clinical indications, fields of application and outcomes. Neurosurg Rev 2025; 48:335. [PMID: 40148698 DOI: 10.1007/s10143-025-03484-3] [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: 11/10/2024] [Revised: 02/20/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
γ-Glutamyl Hydroxymethyl Rhodamine Green (γ-GHRG) fluorescence is a novel fluorescent agent used to enhance tissue visualization during surgical resections, especially in oncological procedures. This systematic review aims to evaluate the efficacy of γ-GHRG fluorescence in improving tumor margin detection, reducing recurrence rates, and enhancing surgical precision. A comprehensive search was conducted across PubMed, Embase, and Cochrane databases up to February 2024, following PRISMA guidelines. A total of 23 studies investigating the use of γ-GHRG fluorescence in surgical resection were included. Data on tumor visualization, surgical margin detection, and postoperative outcomes were analyzed. The included studies demonstrated that γ-GHRG fluorescence significantly improved tumor visualization in a variety of cancers, including ovarian cancer (26.1%), breast cancer (8.7%), lung cancer (8.7%), colorectal cancer (8.7%), kidney cancer (8.7%), head and neck cancer (8.7%), esophageal and gastric cancer (8.7%), prostate cancer (8.7%), brain tumors (gliomas) (4.3%), and liver cancer (4.3%). The probe's high specificity for γ-glutamyl transpeptidase (GGT), which is overexpressed in cancer cells, enabled real-time visualization of tumor margins, allowing for more precise resections. Studies also reported shorter surgical times and lower recurrence rates, particularly in high-grade tumors such as gliomas (4.3%). Although the results are promising, issues related to false positives, tissue specificity, and long-term safety were noted. γ-GHRG fluorescence shows significant potential in enhancing surgical outcomes by providing real-time guidance during tumor resections. Its high specificity for GGT and rapid fluorescence activation make it a valuable tool in cancer surgery. However, further clinical studies are required to address challenges related to sensitivity, specificity, and long-term safety, as well as to explore its application across different cancer types and surgical settings.
Collapse
Affiliation(s)
- Salvatore Marrone
- Department of Neurosurgery, Sant'Elia Hospital, Caltanissetta, Italy
| | - Gianluca Scalia
- Neurosurgery Unit, Highly Specialized Hospital of National Importance "Garibaldi", Catania, Italy.
- Department of Medicine and Surgery, University of Enna "Kore", Enna, Italy.
| | - Oday Atallah
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Giuseppe Scibilia
- Department of Medicine and Surgery, University of Enna "Kore", Enna, Italy
| | - Paolo Palmisciano
- Department of Neurological Surgery, University of California, Davis, Sacramento, CA, USA
| | | | - Rosario Maugeri
- Department of Experimental Biomedicine and Clinical Neurosciences, School of Medicine, Postgraduate Residency Program in Neurological Surgery, Neurosurgical Clinic, AOUP ''Paolo Giaccone'', Palermo, Italy
| | - Domenico Gerardo Iacopino
- Department of Experimental Biomedicine and Clinical Neurosciences, School of Medicine, Postgraduate Residency Program in Neurological Surgery, Neurosurgical Clinic, AOUP ''Paolo Giaccone'', Palermo, Italy
| | - Francesca Graziano
- Neurosurgery Unit, Highly Specialized Hospital of National Importance "Garibaldi", Catania, Italy
| | | | - Ottavio S Tomasi
- Department of Neurosurgery, AUVA Unfallkrankenhaus, Salzburg, Austria
| | - Jessica Naimo
- Pain Therapy and Palliative Care Unit, ASP 7 Ragusa, Ragusa, Italy
| | - Gianluca Ferini
- Department of Medicine and Surgery, University of Enna "Kore", Enna, Italy
- Department of Radiation Oncology, REM Radioterapia Srl, Viagrande, Italy
| | - Paolo Scollo
- Department of Medicine and Surgery, University of Enna "Kore", Enna, Italy
| | - Giuseppe Emmanuele Umana
- Department of Medicine and Surgery, University of Enna "Kore", Enna, Italy
- Department of Neurosurgery, Trauma Center, Gamma Knife Center, Cannizzaro Hospital, Catania, Italy
| |
Collapse
|
2
|
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
|
3
|
Chen K, Teng X, Zhou N, Cheng W. Rising sun or strangled in the cradle? A narrative review of near-infrared fluorescence imaging-guided surgery for pancreatic tumors. Int J Surg 2024; 110:7929-7947. [PMID: 38768476 PMCID: PMC11634182 DOI: 10.1097/js9.0000000000001676] [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: 11/26/2023] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Near-infrared fluorescence (NIRF)-guided surgical navigation has become a promising and effective detection method in pancreatic tumor surgery. The imaging technique has gradually transitioned from the NIR-I region to the NIR-II region. Real-time assessment of the tumor boundary and determination of the ideal resection plane are essential for preserving the pancreatic parenchyma and its secretory functions. However, since the pancreatic parenchyma has a less rich blood supply than the liver, the application of contrast agents in pancreatic tumor surgery is still in its infancy. The application of indocyanine green (ICG) and methylene blue (MB) in intraoperative NIRF imaging of pancreatic tumors has become more mature, but due to the characteristics of nonspecific imaging, the imaging efficiency and depth need to be improved. Many tumor-specific imaging agents have been designed, but most of them have not gone past animal trials because of their high development and imaging costs, biotoxicity, and other limitations. In this article, we review recent reports of ICG, MB, and newly developed contrast agents and imaging devices. We focus on the current status and new developments in the application of these contrast agents and summarize the current clinical and preclinical studies on specific contrast agents. We synthesize relevant reports to discuss the difficulties and prospects of the application of fluorescent imaging agents in pancreatic tumors. We hope that reviewing previous studies and the current progress on contrast imaging technology will provide new perspectives for its future application and development in pancreatic tumor surgery, which should translate into better patient prognoses. The manuscript was written according to the Scale for the Assessment of Narrative Review Articles (SANRA).
Collapse
Affiliation(s)
- Kang Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha
| | - Xiong Teng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Ning Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha
| |
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
|
Cox KE, Turner MA, Lwin TM, Amirfakhri S, Kelly KJ, Hosseini M, Ghosh P, Obonyo M, Hoffman RM, Yazaki PJ, Bouvet M. Targeting Patient-Derived Orthotopic Gastric Cancers with a Fluorescent Humanized Anti-CEA Antibody. Ann Surg Oncol 2024; 31:6291-6299. [PMID: 38888861 PMCID: PMC11300635 DOI: 10.1245/s10434-024-15570-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Gastric cancer poses a major diagnostic and therapeutic challenge as surgical resection provides the only opportunity for a cure. Specific labeling of gastric cancer could distinguish resectable and nonresectable disease and facilitate an R0 resection, which could improve survival. METHODS Two patient-derived gastric cancer lines, KG8 and KG10, were established from surgical specimens of two patients who underwent gastrectomy for gastric adenocarcinoma. Harvested tumor fragments were implanted into the greater curvature of the stomach to establish patient-derived orthotopic xenograft (PDOX) models. M5A (humanized anti-CEA antibody) or IgG control antibodies were conjugated with the near-infrared dye IRDye800CW. Mice received 50 µg of M5A-IR800 or 50 µg of IgG-IR800 intravenously and were imaged after 72 hr. Fluorescence imaging was performed by using the LI-COR Pearl Imaging System. A tumor-to-background ratio (TBR) was calculated by dividing the mean fluorescence intensity of the tumor versus adjacent stomach tissue. RESULTS M5A-IR800 administration resulted in bright labeling of both KG8 and K10 tumors. In the KG8 PDOX models, the TBR for M5A-IR800 was 5.85 (SE ± 1.64) compared with IgG-IR800 at 0.70 (SE ± 0.17). The K10 PDOX models had a TBR of 3.71 (SE ± 0.73) for M5A-IR800 compared with 0.66 (SE ± 0.12) for IgG-IR800. CONCLUSIONS Humanized anti-CEA (M5A) antibodies conjugated to fluorescent dyes provide bright and specific labeling of gastric cancer PDOX models. This tumor-specific fluorescent antibody is a promising potential clinical tool to detect the extent of disease for the determination of resectability as well as to visualize tumor margins during gastric cancer resection.
Collapse
Affiliation(s)
- Kristin E Cox
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Michael A Turner
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Thinzar M Lwin
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Kaitlyn J Kelly
- Department of Surgical Oncology, University of Wisconsin, Madison, WI, USA
| | - Mojgan Hosseini
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marygorret Obonyo
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- AntiCancer Inc, San Diego, CA, USA
| | - Paul J Yazaki
- Department of Immunology & Theranostics, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, La Jolla, CA, USA.
- VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
6
|
Liu Z, Ali M, Sun Q, Zhang Q, Wei C, Wang Y, Tang D, Li X. Current status and future trends of real-time imaging in gastric cancer surgery: A literature review. Heliyon 2024; 10:e36143. [PMID: 39253259 PMCID: PMC11381608 DOI: 10.1016/j.heliyon.2024.e36143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/23/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
Technological advances are crucial for the optimization of gastric cancer surgery, and the success of any gastric cancer surgery is based on the correct and precise anatomical determination of the primary tumour and tissue structures. Real-time imaging-guided surgery is showing increasing potential and utility, mainly because it helps to aid intraoperative decision-making. However, intraoperative imaging faces many challenges in the field of gastric cancer. This article summarizes and discusses the following clinical applications of real-time optical imaging and fluorescence-guided surgery for gastric cancer: (1) the potential of quantitative fluorescence imaging in assessing tissue perfusion, (2) vascular navigation and determination of tumour margins, (3) the advantages and limitations of lymph node drainage assessment, and (4) identification of peritoneal metastases. In addition, preclinical study of tumour-targeted fluorescence imaging are discussed.
Collapse
Affiliation(s)
- Zhu Liu
- The Yangzhou Clinical Medical College of Nanjing Medical University, Yangzhou, 225001, China
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Muhammad Ali
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Qiannan Sun
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Qi Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Chen Wei
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Yong Wang
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Dong Tang
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China
- Yangzhou Key Laboratory of Basic and Clinical Transformation of Digestive and Metabolic Diseases, China
| | - Xin Li
- Northern Jiangsu People's Hospital, Yangzhou, 225001, China
- Department of Pharmacy, Clinical Medical College, Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
7
|
Luo Q, Teng X, Dai M, Yang J, Cheng W, Chen K, Zhou L. Global trends in the application of fluorescence imaging in pancreatic diseases: a bibliometric and knowledge graph analysis. Front Oncol 2024; 14:1383798. [PMID: 39099697 PMCID: PMC11294181 DOI: 10.3389/fonc.2024.1383798] [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: 02/10/2024] [Accepted: 07/01/2024] [Indexed: 08/06/2024] Open
Abstract
Background In recent years, with the continuous development of fluorescence imaging technology, research on its application in pancreatic diseases has surged. This area is currently of high research interest and holds the potential to become a non-invasive and effective tool in the diagnosis and treatment of pancreatic diseases. The objective of this study is to explore the hotspots and trends in the field of fluorescence imaging technology applications in pancreatic diseases from 2003 to 2023 through bibliometric and visual analysis. Methods This study utilized the Web of Science (core collection) to identify publications related to the application of fluorescence imaging technology in pancreatic diseases from 2003 to 2023. Tools such as CiteSpace (V 6.2.R6), VOSviewer (v1.6.20), and R Studio (Bibliometrix: R-tool version 4.1.4) were employed to analyze various dimensions including publication count, countries, institutions, journals, authors, co-cited references, keywords, burst words, and references. Results A comprehensive analysis was conducted on 913 papers published from January 1, 2003, to December 1, 2023, on the application of fluorescence imaging technology in pancreatic diseases. The number of publications in this field has rapidly increased, with the United States being the central hub. The University of California, San Diego emerged as the most active institution. "Biomaterials" was identified as the most influential journal. Authors with the most publications and the highest average citations per article are Hoffman, Robert M. and Luiken, George A., respectively. Keywords such as pancreatic cancer, cancer, expression, indocyanine green, and nanoparticles received widespread attention, with indocyanine green and nanoparticles being current active research hotspots in the field. Conclusion This study is the first bibliometric analysis in the field of fluorescence imaging technology applications in pancreatic diseases. Our data will facilitate a better understanding of the developmental trends, identification of research hotspots, and direction in this field. The findings provide practical information for other scholars to grasp key directions and cutting-edge insights.
Collapse
Affiliation(s)
- Quanneng Luo
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Xiong Teng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - ManXiong Dai
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Jun Yang
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Hunan Schistosomiasis Control Center (Hunan Third People’s Hospital), Yueyang, Hunan, China
| | - Kang Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Lei Zhou
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| |
Collapse
|
8
|
Pringle TA, Chan CD, Luli S, Blair HJ, Rankin KS, Knight JC. Synthesis and In Vivo Evaluation of a Site-specifically Labeled Radioimmunoconjugate for Dual-Modal (PET/NIRF) Imaging of MT1-MMP in Sarcomas. Bioconjug Chem 2022; 33:1564-1573. [PMID: 35867034 PMCID: PMC9389524 DOI: 10.1021/acs.bioconjchem.2c00306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Bone sarcomas are devastating primary bone cancers that
mostly
affect children, young adults, and the elderly. These aggressive tumors
are associated with poor survival, and surgery remains the mainstay
of treatment. Surgical planning is increasingly informed by positron
emission tomography (PET), and tumor margin identification during
surgery is aided by near-infrared fluorescence (NIRF) imaging, yet
these investigations are confounded by probes that lack specificity
for sarcoma biomarkers. We report the development of a dual-modal
(PET/NIRF) immunoconjugate ([89Zr]Zr-DFO-anti-MT1-MMP-IRDye800CW)
that targets MT1-MMP, a matrix metalloproteinase overexpressed in
high-grade sarcomas. [89Zr]Zr-DFO-anti-MT1-MMP-IRDye800CW
was synthesized via site-specific chemoenzymatic
glycan modification, characterized, and isolated in high specific
activity and radiochemical purity. Saturation binding and immunoreactivity
assays indicated only minor perturbation of binding properties. A
novel mouse model of dedifferentiated chondrosarcoma based on intrafemoral
inoculation of HT1080 WT or KO cells (high and low MT1-MMP expression,
respectively) was used to evaluate target binding and biodistribution.
Fluorescence and Cerenkov luminescence images of [89Zr]Zr-DFO-anti-MT1-MMP-IRDye800CW
showed preferential uptake in HT1080 WT tumors. Ex vivo gamma counting revealed that uptake in MT1-MMP-positive tumors was
significantly higher than that in control groups. Taken together,
[89Zr]Zr-DFO-anti-MT1-MMP-IRDye800CW is a promising dual-modal
sarcoma imaging agent for pre-operative surgical planning and intraoperative
surgical guidance.
Collapse
Affiliation(s)
- Toni A Pringle
- School of Natural and Environmental Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K
| | - Corey D Chan
- North of England Bone and Soft Tissue Tumour Service, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Freeman Road, Newcastle Upon Tyne NE7 7DN, U.K.,Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K
| | - Saimir Luli
- Preclinical In Vivo Imaging, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne NE2 4HH, U.K
| | - Helen J Blair
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K.,Wolfson Childhood Cancer Research Centre, Newcastle Upon Tyne NE1 7RY, U.K
| | - Kenneth S Rankin
- North of England Bone and Soft Tissue Tumour Service, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Freeman Road, Newcastle Upon Tyne NE7 7DN, U.K.,Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K
| | - James C Knight
- School of Natural and Environmental Sciences, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K.,Newcastle Centre for Cancer, Newcastle University, Newcastle Upon Tyne NE1 7RU, U.K
| |
Collapse
|
9
|
Abstract
The translation of laboratory science into effective clinical cancer therapy is gaining momentum more rapidly than any other time in history. Understanding cancer cell-surface receptors, cancer cell growth, and cancer metabolic pathways has led to many promising molecular-targeted therapies and cancer gene therapies. These same targets may also be exploited for optical imaging of cancer. Theoretically, any antibody or small molecule targeting cancer can be labeled with bioluminescent or fluorescent agents. In the laboratory setting, fluorescence imaging (FI) and bioluminescence imaging (BLI) have long been used in preclinical research for quantification of tumor bulk, assessment of targeting of tumors by experimental agents, and discrimination between primary and secondary effects of cancer treatments. Many of these laboratory techniques are now moving to clinical trials. Imageable engineered fluorescent probes that are highly specific for cancer are being advanced. This will allow for the identification of tumors for staging, tracking novel therapeutic agents, assisting in adequate surgical resection, and allowing image-guided biopsies. The critical components of FI include (1) a fluorescent protein that is biologically safe, stable, and distinctly visible with a high target to background ratio and (2) highly sensitive optical detectors. This review will summarize the most promising optical imaging agents and detection devices for cancer clinical research and clinical care.
Collapse
|
10
|
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
|
11
|
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
|
12
|
Lwin TM, Turner MA, Amirfakhri S, Nishino H, Debie P, Cosman BC, Hoffman RM, Hernot S, Bouvet M. Rapid tumor-labeling kinetics with a site-specific near-infrared anti-CEA nanobody in a patient-derived orthotopic xenograft mouse model of colon cancer. J Surg Oncol 2021; 124:1121-1127. [PMID: 34309885 PMCID: PMC8556245 DOI: 10.1002/jso.26623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND/OBJECTIVES Nanobodies are the smallest biologic antigen-binding fragments derived from camelid-derived antibodies. Nanobodies effect a peak tumor signal within minutes of injection and present a novel opportunity for fluorescence-guided surgery (FGS). The present study demonstrates the efficacy of an anti-CEA nanobody conjugated to near-infrared fluorophore LICOR-IRDye800CW for rapid intraoperative tumor labeling of colon cancer. METHODS LS174T human colon cancer cells or fragments of patient-derived colon cancer were implanted subcutaneously or orthotopically in nude mice. Anti-CEA nanobodies were conjugated with IRDye800CW and 1-3 nmol were injected intravenously. Mice were serially imaged over time. Peak fluorescence signal and tumor-to-background ratio (TBR) were recorded. RESULTS Colon cancer tumors were detectable using fluorescent anti-CEA nanobody within 5 min of injection at all three doses. Maximal fluorescence intensity was observed within 15 min-3 h for all three doses with TBR values ranging from 1.3 to 2.3. In the patient-derived model of colon cancer, fluorescence was detectable with a TBR of 4.6 at 3 h. CONCLUSIONS Fluorescent anti-CEA nanobodies rapidly and specifically labeled colon cancer in cell-line-based and patient-derived orthotopic xenograft (PDOX) models. The kinetics of nanobodies allow for same day administration and imaging. Anti-CEA-nb-800 is a promising and practical molecule for FGS of colon cancer.
Collapse
Affiliation(s)
- Thinzar M. Lwin
- Department of Surgery, University of California San Diego, San Diego, California, USA
- Department of Surgical Oncology, Dana Farber Cancer Center, Boston, Massachusetts, USA
| | - Michael A. Turner
- Department of Surgery, University of California San Diego, San Diego, California, USA
- VA San Diego Healthcare System, San Diego, California, USA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, San Diego, California, USA
- VA San Diego Healthcare System, San Diego, California, USA
| | - Hiroto Nishino
- Department of Surgery, University of California San Diego, San Diego, California, USA
- VA San Diego Healthcare System, San Diego, California, USA
| | - Pieterjan Debie
- Laboratory for In vivo Cellular and Molecular Imaging (ICMI-BEFY-MIMA), Vrije Universiteit Brussel, Brussels, Belgium
| | - Bard C. Cosman
- Department of Surgery, University of California San Diego, San Diego, California, USA
- VA San Diego Healthcare System, San Diego, California, USA
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, San Diego, California, USA
- VA San Diego Healthcare System, San Diego, California, USA
- AntiCancer, Inc., San Diego, California, USA
| | - Sophie Hernot
- Laboratory for In vivo Cellular and Molecular Imaging (ICMI-BEFY-MIMA), Vrije Universiteit Brussel, Brussels, Belgium
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, California, USA
- VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
13
|
Choi JH, Park JY. Insulin-Like Growth Factor-1 Receptor Targeted Fluorescent Imaging for Gallbladder Cancer in Orthotopic Mouse Models. Gut Liver 2021; 16:606-612. [PMID: 34462395 PMCID: PMC9289833 DOI: 10.5009/gnl210164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/22/2021] [Accepted: 07/13/2021] [Indexed: 11/06/2022] Open
Abstract
Background/Aims Gallbladder cancer is fatal, but fluorescent imaging technology can facilitate timely diagnosis and improve patient outcomes. Fluorophore-conjugated insulin-like growth factor-1 receptor (IGF-1R) targeted antibodies were used to visualize gallbladder cancer in orthotopic tumor mouse models. Methods Western blotting, flow cytometric analysis, and confocal microscopy detected the expression of IGF-1R in SNU-308, SNU-478, and SNU-1196 bile duct cancer cells. In vivo imaging of SNU-478 and SNU-1196 subcutaneous tumors and orthotopic gallbladder tumor models of SNU-478 were performed after injection with DyLight 650-conjugated IGF-1R antibody. Results Western blotting and flow cytometric analysis showed that IGF-1R was expressed in bile duct cancer cells, and confocal microscopy demonstrated that IGF-1R antibody was able to bind to IGF-1R on the cell membrane. Fluorescent IGF-1R antibody injected into the mouse tail vein made subcutaneous tumors and orthotopic tumors become fluorescent. The intensity of fluorescence from the tumor was stronger than that from surrounding normal tissues. Histochemical examination confirmed that the tumor was located inside the gallbladder and adjacent liver parenchyma of mice. Conclusions Our study showed that a fluorescent IGF-1R-targeted antibody could help detect gallbladder tumors. Tumor-specific imaging technology can be applied to endoscopy, laparoscopy, and robotic surgery for better management of gallbladder cancer.
Collapse
Affiliation(s)
- Jung Ha Choi
- Division of Gastroenterology, Department of Internal Medicine and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Hollandsworth HM, Turner MA, Hoffman RM, Bouvet M. A review of tumor-specific fluorescence-guided surgery for colorectal cancer. Surg Oncol 2021; 36:84-90. [PMID: 33316684 PMCID: PMC7855598 DOI: 10.1016/j.suronc.2020.11.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 11/18/2020] [Accepted: 11/27/2020] [Indexed: 01/08/2023]
Abstract
The present study reviews the use of tumor-specific antibodies conjugated to fluorescent dyes in preclinical and clinical studies to enhance visualization of primary tumors and metastases for fluorescence-guided surgery (FGS) in colorectal cancer (CRC). A search strategy was developed using the peer-reviewed National Center for Biotechnology Information (NCBI) database on PubMed. Studies using tumor-specific fluorescence imaging and FGS techniques on murine models of colorectal cell lines or patient-derived orthotopic xenograft (PDOX) colorectal cancer are reviewed. A total of 24 articles were identified that met the inclusion criteria, 21 preclinical and 3 clinical trials. The most widely used target antigen in preclinical and clinical trials was carcinoembryonic antigen (CEA). Mouse studies and clinical studies have demonstrated that the use of FGS in CRC can aid in decreased residual tumor and decreased rates of recurrence. As the mainstay of colorectal cancer treatment is surgery, the addition of intraoperative fluorescence imaging can help locate tumor margins, visualize occult micro-metastases, drive surgical decision making and improve patient outcomes.
Collapse
Affiliation(s)
- Hannah M Hollandsworth
- Department of Surgery, University of California San Diego, San Diego, CA, USA; Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - Michael A Turner
- Department of Surgery, University of California San Diego, San Diego, CA, USA; Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, San Diego, CA, USA; Moores Cancer Center, University of California San Diego, San Diego, CA, USA; AntiCancer Inc., San Diego, CA, USA; VA San Diego Healthcare System, San Diego, CA, USA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, CA, USA; Moores Cancer Center, University of California San Diego, San Diego, CA, USA; VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
15
|
Unique Benefits of Tumor-Specific Nanobodies for Fluorescence Guided Surgery. Biomolecules 2021; 11:biom11020311. [PMID: 33670740 PMCID: PMC7921980 DOI: 10.3390/biom11020311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor-specific fluorescence labeling is promising for real-time visualization of solid malignancies during surgery. There are a number of technologies to confer tumor-specific fluorescence. Antibodies have traditionally been used due to their versatility in modifications; however, their large size hampers efficient fluorophore delivery. Nanobodies are a novel class of molecules, derived from camelid heavy-chain only antibodies, that have shown promise for tumor-specific fluorescence labeling. Nanobodies are ten times smaller than standard antibodies, while maintaining antigen-binding capacity and have advantageous features, including rapidity of tumor labeling, that are reviewed in the present report. The present report reviews special considerations needed in developing nanobody probes, the status of current literature on the use of nanobody probes in fluorescence guided surgery, and potential challenges to be addressed for clinical translation.
Collapse
|
16
|
Huang A, Fan W, Liu J, Huang B, Cheng Q, Wang P, Duan Y, Ma T, Chen L, Wang Y, Yu M. Prognostic Role of S100A8 in Human Solid Cancers: A Systematic Review and Validation. Front Oncol 2020; 10:564248. [PMID: 33240811 PMCID: PMC7682514 DOI: 10.3389/fonc.2020.564248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
Background S100A8 plays a key role in many cellular processes and is highly expressed in various solid cancers. However, the prognostic role of S100A8 has not been well defined. Therefore, we conducted a quantitative meta-analysis to investigate whether or not S100A8 could be used as a prognostic biomarker in solid tumors. Methods PubMed, Web of Science, Embase, and Cochrane library were searched to acquire relevant studies that evaluated the association between expression of S100A8 and prognosis of cancer patients. Pooled hazard ratios (HRs) with their corresponding 95% confidence intervals (CIs) were extracted to evaluate the association between S100A8 overexpression and Overall Survival (OS), Disease-Free Survival (DFS), Recurrence-Free Survival (RFS), and Progression-Free Survival (PFS). The expression of S100A8 was also validated by Flow cytometry, immunohistochemistry (IHC), and western blot. Results A total of 2,817 patients from 13 independent studies, ranging from 43 to 1,117 patients in size, were statistically analyzed. Our results indicated that a high level of S100A8 expression was significantly associated with poor OS, poor DFS, and poor PFS/RFS. In term of clinical pathological characteristics, a high expression level of S100A8 was significantly associated with differentiation grades, lymphatic metastasis, ER statue, and PR statue. The validation studies showed that the expression of S100A8 was at high levels in MDA-MB-231 (79.7%), MDA-MB-453 (89.2%), HTB-9 (70.2%), and T24 (53.3%) cells and it was higher in breast cancer tissue and bladder cancer tissue than their corresponding para-carcinoma tissue. Conclusions S100A8 overexpression was significantly associated with poor clinical prognosis in cancer patients. S100A8 is potential a prognostic biomarker in breast cancer and bladder cancer. More well-designed studies with adequate prognostic data are needed to confirm the prognostic role of S100A8 revealed in this study.
Collapse
Affiliation(s)
- An Huang
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China.,Key Laboratory of Carcinogenesis andTranslational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Fan
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiacui Liu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ben Huang
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qingyuan Cheng
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ping Wang
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yiping Duan
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tiantian Ma
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Liangyue Chen
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanping Wang
- Department of Obstetrics and Gynecology, Hubei Provincial Hospital of TCM, Wuhan, China
| | - Mingxia Yu
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Lwin TM, Hernot S, Hollandsworth H, Amirfakhri S, Filemoni F, Debie P, Hoffman RM, Bouvet M. Tumor-specific near-infrared nanobody probe rapidly labels tumors in an orthotopic mouse model of pancreatic cancer. Surgery 2020; 168:85-91. [PMID: 32370916 DOI: 10.1016/j.surg.2020.02.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/07/2020] [Accepted: 02/18/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Nanobodies, derived from camelid antibodies made of only heavy chains, are the smallest, biologic, antigen-binding fragments (~15kDa) with faster pharmacokinetics and better tumor penetration efficiency than standard antibodies. The present study evaluates the efficacy of a fluorescent, anti-carcinoembryonic antigen (CEA) nanobody for rapid tumor labeling in an orthotopic mouse model of pancreatic cancer. METHODS Anti-CEA or control nanobodies were conjugated with the near-infrared fluorophore IRDye 800CW. Fragments of BxPC-3 (high-CEA expressing) or MiaPACA-2 (low-CEA expressing) human pancreatic cancer cell lines were orthotopically implanted into the pancreatic tail of nude mice. After tumors reached 7 to 10 mm in size, 2 nmol anti-CEA or control nanobody-IRDye800CW were injected intravenously. Mice were imaged at various time points hours post-injection. RESULTS Anti-CEA nanobodies clearly labeled BxPC3 orthotopic pancreatic tumors 3 hours after injection. The signal was present as early as 15 minutes after injection and was robust at 1 to 3 hours after injection with a tumor-to-background ratio of 2.66. In contrast, there was very low accumulation in the low CEA-expressing, MiaPACA2 pancreatic orthotopic tumors. The fluorophore-conjugated nanobody was specific for CEA-expressing tumors, while the control nanobody did not show any tumor-specific signal. Both nanobodies had strong kidney uptake as expected for small-molecule probes. The fluorescence signal was detectable using 2 clinical, Food and Drug Administration-approved, 800 nm imaging devices as well as small animal imaging systems. CONCLUSION This anti-CEA, nanobody-based, fluorescent probe labeled pancreatic orthotopic tumors within 15 minutes of intravenous injection. Fluorescent anti-CEA nanobodies have labeling kinetics that approach the speed of nonspecific dyes such as indocyanine green but with the specificity of antibodies. The use of fluorescently-labeled, intact antibodies leads to a labeling delay of 48 to 96 hours between probe administration and the necessarily delayed time of operation, which can be avoided with nanobodies. The kinetics of a nanobody-based probe makes it a practical agent for same-day, patient administration and fluorescence-guided surgery.
Collapse
Affiliation(s)
- Thinzar M Lwin
- Department of Surgery, University of California San Diego, CA
| | - Sophie Hernot
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hannah Hollandsworth
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA
| | - Filemoni Filemoni
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA
| | - Pieterjan Debie
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Brussels, Belgium
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA; AntiCancer, Inc, San Diego, CA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, CA; VA San Diego Healthcare System, CA.
| |
Collapse
|
18
|
Kose E, Kahramangil B, Aydin H, Donmez M, Takahashi H, Acevedo-Moreno LA, Sasaki K, Aucejo F, Berber E. A comparison of indocyanine green fluorescence and laparoscopic ultrasound for detection of liver tumors. HPB (Oxford) 2020; 22:764-769. [PMID: 31653594 DOI: 10.1016/j.hpb.2019.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 08/12/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Indocyanine green (ICG) fluorescence imaging (ICG-FI) has been suggested for intraoperative identification of liver tumors. We aim to compare the intraoperative diagnostic utility of this imaging modality with laparoscopic ultrasound (LUS). METHODS This is an IRB-approved prospective study. ICG was administered intravenously 1-2 days before surgery. The findings on ICG-FI were compared to those on preoperative cross-sectional imaging (POCSI), LUS, diagnostic laparoscopy (DL). RESULTS A total of 144 lesions (62 superficial [visible on DL] and 82 deep) were detected in the study patients. POCSI identified 74%, LUS identified 92%, and ICG-FI identified 43%. ICG-FI detection rate was higher for superficial (95%) versus deep lesions (4%). 3% (4/144) of all lesions were seen only on ICG-FI. However, all of these lesions were small and superficial lesions that were apparent on DL. CONCLUSION Although ICG-FI allowed detection of small superficial lesions that were not identifiable by POCSI or LUS, these lesions were apparent on DL even before ICG-FI. Therefore, its utility as an intraoperative diagnostic modality is limited at the dosage and timing used in the study. We believe that rather than a diagnostic tool, it has more potential for a dynamic use in guiding the resection of superficial lesions and delineating segmental/lobar anatomy.
Collapse
Affiliation(s)
- Emin Kose
- Department of Endocrine Surgery, Cleveland Clinic, OH, United States
| | - Bora Kahramangil
- Department of Endocrine Surgery, Cleveland Clinic, OH, United States
| | - Husnu Aydin
- Department of Endocrine Surgery, Cleveland Clinic, OH, United States
| | - Mustafa Donmez
- Department of Endocrine Surgery, Cleveland Clinic, OH, United States
| | - Hideo Takahashi
- Department of General Surgery, Cleveland Clinic, OH, United States
| | | | - Kazunari Sasaki
- Department of General Surgery, Cleveland Clinic, OH, United States
| | - Federico Aucejo
- Department of General Surgery, Cleveland Clinic, OH, United States
| | - Eren Berber
- Department of Endocrine Surgery, Cleveland Clinic, OH, United States; Department of General Surgery, Cleveland Clinic, OH, United States.
| |
Collapse
|
19
|
Anti-carcinoembryonic antigen-related cell adhesion molecule antibody for fluorescence visualization of primary colon cancer and metastases in patient-derived orthotopic xenograft mouse models. Oncotarget 2020; 11:429-439. [PMID: 32064046 PMCID: PMC6996915 DOI: 10.18632/oncotarget.27446] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/04/2020] [Indexed: 01/30/2023] Open
Abstract
Background: Monoclonal antibody (mAb) 6G5j is a novel anti-CEACAM monoclonal antibody. Our aim was to investigate mAb 6G5j binding characteristics and to validate fluorescence targeting of colorectal tumors and metastases in patient derived orthotopic xenograft (PDOX) models with fluorescently labeled 6G5j. Materials/Methods: The MAb 6G5j binding profile was analyzed with ELISA, Western blot and immunohistochemistry. MAb 6G5j was conjugated to near-infrared dye IR800CW (LI-COR). Western blotting was performed with various colon cancer cell lysates to determine CEACAM expression. Nude mice received orthotopic implantation of patient-derived primary colon cancer and patient-derived colon cancer metastases. Mice were administered varying doses of 6G5j-IR800CW via tail vein injection and imaged 24 and 48 hours later. Results: MAb 6G5j bound to human CEACAM1, 3, 5, 6 and 8. Western blotting demonstrated varied expression of CEACAMs in 15 of 16 colon cancer lysates. Dose and time-response imaging demonstrated optimal imaging 48 hours after administration of 50 μg 6G5j-IR800CW (Tumor-to-liver ratio (TLR) 3.17, SEM ± 0.45). Primary cancers and multiple metastases were fluorescently visualized. Conclusions: Anti-CEACAM antibody 6G5j binds multiple CEACAMs which may lead to improved detection of tumor margins for tumors and metastases that have variable expression of CEA and other CEACAMs. 6G5j mAb may be useful for colon cancer detection for pre-surgical diagnosis and fluorescence-guided surgery.
Collapse
|
20
|
Pèlegrin A, Gutowski M, Cailler F. Les anticorps, outils de choix pour la chirurgie guidée par fluorescence. Med Sci (Paris) 2020; 35:1066-1071. [DOI: 10.1051/medsci/2019207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
La chirurgie guidée par fluorescence se développe en clinique depuis plusieurs années. Si l’utilisation de colorants non ciblés peut être utile dans certaines pathologies, des agents de contraste spécifiques sont indispensables en oncologie. Comme le montrent les dernières études cliniques, les anticorps monoclonaux ont toutes les caractéristiques pour jouer un rôle majeur dans ce domaine d’imagerie médicale, à condition que la cible antigénique soit pertinente.
Collapse
|
21
|
Zian W, Yang L, Peng W, Yifei J, Min J. Small molecular interaction-based fluorescence enhancement for second near-infrared imaging. Nanomedicine (Lond) 2020; 15:115-129. [DOI: 10.2217/nnm-2019-0233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: This study described a new strategy to enhance second near-infrared (NIR-II) fluorescence intensity. Materials & methods: NIR-II liposomes were prepared by thin film hydration method and their fluorescence properties were evaluated. The efficacy of the optimized liposome was then evaluated in vivo with low dose and irradiation. Results: Indocyanine green-IR1061 liposome exhibited higher fluorescence intensity (∼fourfold than IR1061 liposome) with the red-shifted emission. The intensity of indocyanine green-IR1061 cationic liposome was enhanced to approximately tenfold, which allowed us to perform angiography with lower doses and less exposure time. Conclusion: We report a new and efficient way to enhance NIR-II fluorescence intensity. This could be used to acquire high temporal resolution and signal-to-background ratio fluorescence imaging.
Collapse
Affiliation(s)
- Wang Zian
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Liu Yang
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Wang Peng
- Stake Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China
- Department of Biomedical Engineering,School of Engineering, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jiang Yifei
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Ji Min
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, PR China
| |
Collapse
|
22
|
Picchetto A, Seeliger B, La Rocca S, Barberio M, D'Ambrosio G, Marescaux J, Diana M. [Fluorescence-guided detection of lymph node metastases of gastrointestinal tumors]. Chirurg 2019; 90:891-898. [PMID: 31552436 DOI: 10.1007/s00104-019-01039-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A correct lymph node (LN) staging is essential in oncological surgery. Indocyanine green (ICG) near-infrared fluorescence (NIRF) guided sentinel lymph node (SLN) navigation is a relatively novel technique. The aim of this review is to analyze the impact of ICG-NIRF on identification of LN metastases of gastrointestinal tumors. The Scopus and PubMed/MEDLINE literature databases were searched and 20 studies were included. The ICG-NIRF navigation of LN has been shown to enable and improve LN detection in gastrointestinal tumors; however, the mean detection, sensitivity, accuracy and false negative rates show substantial variation. This could be due to both the heterogeneous techniques applied and to the low retention of ICG by lymph nodes. Fluorescence imaging to identify LN drainage is a promising tool to improve oncological outcomes. Nonetheless, the technique requires further development in terms of hardware, software and fluorophores, which are currently being investigated.
Collapse
Affiliation(s)
- Andrea Picchetto
- General and Colorectal Surgery Division, Department of Cardiothoracic, Vascular Surgery and Organ Transplantation, Policlinico Umberto I University Hospital, Sapienza University of Rome, Viale del Policlinico 155, Rom, Italien.
| | - Barbara Seeliger
- Institute of Image-Guided Surgery, IHU-Strasbourg, Straßburg, Frankreich
- Research Institute against Cancer of the Digestive System, IRCAD, Straßburg, Frankreich
- Kliniken Essen-Mitte, Essen, Deutschland
| | - Stefania La Rocca
- General and Colorectal Surgery Division, Department of Cardiothoracic, Vascular Surgery and Organ Transplantation, Policlinico Umberto I University Hospital, Sapienza University of Rome, Viale del Policlinico 155, Rom, Italien
| | - Manuel Barberio
- Institute of Image-Guided Surgery, IHU-Strasbourg, Straßburg, Frankreich
- Research Institute against Cancer of the Digestive System, IRCAD, Straßburg, Frankreich
| | - Giancarlo D'Ambrosio
- General and Colorectal Surgery Division, Department of Cardiothoracic, Vascular Surgery and Organ Transplantation, Policlinico Umberto I University Hospital, Sapienza University of Rome, Viale del Policlinico 155, Rom, Italien
| | - Jacques Marescaux
- Institute of Image-Guided Surgery, IHU-Strasbourg, Straßburg, Frankreich
- Research Institute against Cancer of the Digestive System, IRCAD, Straßburg, Frankreich
| | - Michele Diana
- Institute of Image-Guided Surgery, IHU-Strasbourg, Straßburg, Frankreich
- Research Institute against Cancer of the Digestive System, IRCAD, Straßburg, Frankreich
| |
Collapse
|
23
|
Recent advances on small-molecule fluorophores with emission beyond 1000 nm for better molecular imaging in vivo. CHINESE CHEM LETT 2019. [DOI: 10.1016/j.cclet.2019.05.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
24
|
Yazaki PJ, Lwin TM, Minnix M, Li L, Sherman A, Molnar J, Miller A, Frankel P, Chea J, Poku E, Bowles N, Hoffman RM, Shively JE, Bouvet M. Improved antibody-guided surgery with a near-infrared dye on a pegylated linker for CEA-positive tumors. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-9. [PMID: 31254333 PMCID: PMC6978469 DOI: 10.1117/1.jbo.24.6.066012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/04/2019] [Indexed: 05/09/2023]
Abstract
Real-time intraoperative image-guided cancer surgery promises to improve oncologic outcomes. Tumor-specific antibodies conjugated with near-infrared (NIR) fluorophores have demonstrated the potential to enhance visualization of solid tumor margins and metastatic disease; however, multiple challenges remain, including improvement in probe development for clinical utility. We have developed an NIR-IR800 dye on a PEGylated linker (sidewinder) conjugated to the humanized anti-carcinoembryonic antigen (CEA) antibody (M5A) with extended in vivo serum and tumor persistence. The anti-CEA M5A-sidewinder has a high dye-to-antibody ratio (average of 7 per antibody) that allows, in an orthotopic implanted human pancreatic cancer mouse model increased tumor fluorescence, higher tumor-to-background ratio and extends the surgical scheduling window compared to current antibody dye conjugates. These preclinical results demonstrate the potential of this probe for fluorescence-guided surgery of CEA-positive gastrointestinal cancers.
Collapse
Affiliation(s)
- Paul J. Yazaki
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
- Address all correspondence to Paul J. Yazaki, E-mail:
| | - Thinzar M. Lwin
- University of California San Diego, Department of Surgery, La Jolla, California, United States
| | - Megan Minnix
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Lin Li
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Anakim Sherman
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Justin Molnar
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Aaron Miller
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Paul Frankel
- Beckman Research Institute, Department of Computational and Quantitative Medicine, City of Hope, Duarte, California, United States
| | - Junie Chea
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Erasmus Poku
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Nicole Bowles
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Robert M. Hoffman
- University of California San Diego, Department of Surgery, La Jolla, California, United States
- AntiCancer, Inc., San Diego, California, United States
- VA San Diego Healthcare System, San Diego, California, United States
| | - John E. Shively
- Beckman Research Institute, Diabetes Metabolism Research Institute, Department of Molecular Imaging and Therapy, City of Hope, Duarte, California, United States
| | - Michael Bouvet
- University of California San Diego, Department of Surgery, La Jolla, California, United States
- VA San Diego Healthcare System, San Diego, California, United States
| |
Collapse
|
25
|
de Souza ALR, Marra K, Gunn J, Samkoe KS, Hull S, Paulsen KD, Pogue BW. Optimizing Glioma Detection Using an EGFR-Targeted Fluorescent Affibody. Photochem Photobiol 2018; 94:1167-1171. [PMID: 30129069 PMCID: PMC6234080 DOI: 10.1111/php.13003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/14/2018] [Indexed: 12/13/2022]
Abstract
Since many types of cancers overexpress EGFR, this surface receptor has been used as a target for therapy or diagnosis of malignant disease. Uptake kinetics of EGFR-targeted fluorescent Affibody (ABY-029) were studied with a view toward optimizing efficacy of tumor detection in a glioma as a function of both delivered dose and concurrent administration of unlabeled cetuximab (an EGFR antagonist). U251 glioma cells were inoculated in brain of nude rats, and the fluorescence from each brain was analyzed after the administration of ABY-029. Although cetuximab was able to systematically block ABY-029 binding to EGFR in a dose-dependent manner in cell culture, no influence on the tumor-to-normal brain contrast was seen when unlabeled cetuximab was administered prior to ABY-029. Ex vivo imaging of ABY-029 fluorescence showed increasing values of the tumor-to-normal brain ratio with an increasing injected dose. A saturation value was obtained at a dose of 245 μg kg-1 which represents a 10-fold increase over a "microdose" value. According to FDA, the microdose of protein products is considered ≤30 nanomoles due to its difference in molecular weight as compared to synthetic drugs. This observation indicates that glioma detection will be optimal if the ABY-029 dose exceeds the "microdose" value.
Collapse
Affiliation(s)
- Ana Luiza Ribeiro de Souza
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- CAPES Foundation, Ministry of Education of Brazil, Brasília 70040-020, Brazil
| | - Kayla Marra
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Jason Gunn
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Kimberley S. Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Department of Surgery, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Sally Hull
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Keith D. Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Department of Surgery, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
- Department of Surgery, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| |
Collapse
|
26
|
Vuijk FA, Hilling DE, Mieog JSD, Vahrmeijer AL. Fluorescent-guided surgery for sentinel lymph node detection in gastric cancer and carcinoembryonic antigen targeted fluorescent-guided surgery in colorectal and pancreatic cancer. J Surg Oncol 2018; 118:315-323. [PMID: 30216455 PMCID: PMC6175076 DOI: 10.1002/jso.25139] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/29/2018] [Indexed: 12/24/2022]
Abstract
Sentinel lymph node procedures for gastric cancer resections using indocyanine green (ICG) linked to Nanocoll outperformed normal ICG but did not provide information on possible lymph node metastasis. Carcinoembryonic antigen targeted fluorescent imaging using SGM‐101 was successful in both pancreatic and colorectal cancer. A large phase III multicentre trial will soon be initiated in colorectal cancer patients.
Collapse
Affiliation(s)
- Floris A Vuijk
- Department of Surgery, Leiden University Medical Centre, Leiden, The Netherlands
| | - Denise E Hilling
- Department of Surgery, Leiden University Medical Centre, Leiden, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Centre, Leiden, The Netherlands
| | | |
Collapse
|
27
|
Hoogstins CES, Boogerd LSF, Sibinga Mulder BG, Mieog JSD, Swijnenburg RJ, van de Velde CJH, Farina Sarasqueta A, Bonsing BA, Framery B, Pèlegrin A, Gutowski M, Cailler F, Burggraaf J, Vahrmeijer AL. Image-Guided Surgery in Patients with Pancreatic Cancer: First Results of a Clinical Trial Using SGM-101, a Novel Carcinoembryonic Antigen-Targeting, Near-Infrared Fluorescent Agent. Ann Surg Oncol 2018; 25:3350-3357. [PMID: 30051369 PMCID: PMC6132431 DOI: 10.1245/s10434-018-6655-7] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Indexed: 01/12/2023]
Abstract
Background Near-infrared (NIR) fluorescence is a promising novel imaging technique that can aid in intraoperative demarcation of pancreatic cancer (PDAC) and thus increase radical resection rates. This study investigated SGM-101, a novel, fluorescent-labeled anti-carcinoembryonic antigen (CEA) antibody. The phase 1 study aimed to assess the tolerability and feasibility of intraoperative fluorescence tumor imaging using SGM-101 in patients undergoing a surgical exploration for PDAC. Methods At least 48 h before undergoing surgery for PDAC, 12 patients were injected intravenously with 5, 7.5, or 10 mg of SGM-101. Tolerability assessments were performed at regular intervals after dosing. The surgical field was imaged using the Quest NIR imaging system. Concordance between fluorescence and tumor presence on histopathology was studied. Results In this study, SGM-101 specifically accumulated in CEA-expressing primary tumors and peritoneal and liver metastases, allowing real-time intraoperative fluorescence imaging. The mean tumor-to-background ratio (TBR) was 1.6 for primary tumors and 1.7 for metastatic lesions. One false-positive lesion was detected (CEA-expressing intraductal papillary mucinous neoplasm). False-negativity was seen twice as a consequence of overlying blood or tissue that blocked the fluorescent signal. Conclusion The use of a fluorescent-labeled anti-CEA antibody was safe and feasible for the intraoperative detection of both primary PDAC and metastases. These results warrant further research to determine the impact of this technique on clinical decision making and overall survival.
Collapse
Affiliation(s)
- Charlotte E S Hoogstins
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.,Centre for Human Drug Research, Leiden, The Netherlands
| | - Leonora S F Boogerd
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | - André Pèlegrin
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | - Marian Gutowski
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | | | - Jacobus Burggraaf
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden Academic Center for Drug Research, Leiden, The Netherlands
| | | |
Collapse
|
28
|
Lwin TM, Hoffman RM, Bouvet M. The development of fluorescence guided surgery for pancreatic cancer: from bench to clinic. Expert Rev Anticancer Ther 2018; 18:651-662. [PMID: 29768067 PMCID: PMC6298876 DOI: 10.1080/14737140.2018.1477593] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Surgeons face major challenges in achieving curative R0 resection for pancreatic cancers. When the lesion is localized, they must appropriately visualize the tumor, determine appropriate resection margins, and ensure complete tumor clearance. Real-time surgical navigation using fluorescence-guidance has enhanced the ability of surgeons to see the tumor and has the potential to assist in achieving more oncologically complete resections. When there is metastatic disease, fluorescence enhancement can help detect these lesions and prevent unnecessary and futile surgeries. Areas covered: This article reviews different approaches for delivery of a fluorescence signal, their pre-clinical and clinical developments for fluorescence guided surgery, the advantages/challenges of each, and their potential for advancements in the future. Expert commentary: A variety of molecular imaging techniques are available for delivering tumor-specific fluorescence signals. Significant advancements have been made in the past 10 years due to the large body of literature on targeted therapies and this has translated into rapid developments of tumor-specific probes.
Collapse
Affiliation(s)
- Thinzar M. Lwin
- Department of Surgery, University of California San Diego, San Diego, CA
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, San Diego, CA
- AntiCancer, Inc., San Diego, CA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, CA
- VA San Diego Healthcare System, San Diego, CA
| |
Collapse
|
29
|
Ding F, Zhan Y, Lu X, Sun Y. Recent advances in near-infrared II fluorophores for multifunctional biomedical imaging. Chem Sci 2018; 9:4370-4380. [PMID: 29896378 PMCID: PMC5961444 DOI: 10.1039/c8sc01153b] [Citation(s) in RCA: 341] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/19/2018] [Indexed: 12/19/2022] Open
Abstract
In recent years, owing to unsatisfactory clinical imaging clarity and depths in the living body for early diagnosis and prognosis, novel imaging modalities with high bioimaging performance have been actively explored. The remarkable headway made in the second near-infrared region (NIR-II, 1000-1700 nm) has promoted the development of biomedical imaging significantly. NIR-II fluorescence imaging possesses a number of merits which prevail over the traditional and NIR-I (400-900 nm) imaging modalities in fundamental research, such as reduced photon scattering, as well as auto-fluorescence and improved penetration depth. Functional probes for instant and precise feedback of in vivo information are at the core of this modality for superb imaging. Herein, we review the recently developed fluorophores including carbon nanotubes, organic small molecules, quantum dots, conjugated polymers and rare-earth-doped materials to present superior and multifunctionality of biomedical imaging in the NIR-II regions (1000-1700 nm).
Collapse
Affiliation(s)
- Feng Ding
- Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , International Joint Research Center for Intelligent Biosensor Technology and Health , Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis , Chemical Biology Center , College of Chemistry , Central China Normal University , Wuhan 430079 , China .
| | - Yibei Zhan
- School of Chemistry and Chemical Engineering , Hubei Polytechnic University , Hubei 435003 , China
| | - Xiaoju Lu
- School of Chemistry and Chemical Engineering , Hubei Polytechnic University , Hubei 435003 , China
| | - Yao Sun
- Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , International Joint Research Center for Intelligent Biosensor Technology and Health , Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis , Chemical Biology Center , College of Chemistry , Central China Normal University , Wuhan 430079 , China .
| |
Collapse
|
30
|
Moore LS, Rosenthal EL, de Boer E, Prince AC, Patel N, Richman JM, Morlandt AB, Carroll WR, Zinn KR, Warram JM. Effects of an Unlabeled Loading Dose on Tumor-Specific Uptake of a Fluorescently Labeled Antibody for Optical Surgical Navigation. Mol Imaging Biol 2018; 19:610-616. [PMID: 27830425 DOI: 10.1007/s11307-016-1022-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE Intraoperative optical imaging to guide surgeons during oncologic resections offers a unique and promising solution to the ambiguity of cancer margins to tactile and visual assessment that results in devastatingly high rates of positive margins. Sequestering of labeled antibodies by normal tissues with high expression of the antibody target, or "antigen sinks", diminishes the efficacy of these probes to provide contrast between the tumor and background tissues by decreasing the amount of circulating probe available for uptake by the tumor and by increasing the fluorescence of non-tumor tissues. We hypothesized that administering a dose of unlabeled antibody prior to infusion of the near-infrared (NIR) fluorescently labeled antibody would improve tumor-specific uptake and contrast of the fluorescently labeled probe by occupying extra-tumoral binding sites, thereby increasing the amount of labeled probe available for uptake by the tumor. PROCEDURES In this study, we explore this concept by testing two different "pre-load" doses of unlabeled cetuximab (the standard 10-mg test dose, and a larger, experimental 100-mg test dose) in six patients receiving cetuximab conjugated to the fluorescent dye IRDye800CW (cetuximab-IRDye800CW) in a clinical trial, and compared the amount of fluorescent antibody in tumor and background tissues, as well as the tumor-specific contrast of each. RESULTS The patients receiving the larger preload (100 mg) of unlabeled cetuximab demonstrated significantly higher concentrations (9.5 vs. 0.1 μg) and a longer half-life (30.3 vs. 20.6 days) of the labeled cetuximab in plasma, as well as significantly greater tumor fluorescence (32.3 vs. 9.3 relative fluorescence units) and tumor to background ratios (TBRs) (5.5 vs. 1.7). CONCLUSIONS Administering a preload of unlabeled antibody prior to infusion of the fluorescently labeled drug may be a simple and effective way to improve the performance of antibody-based probes to guide surgical resection of solid malignancies.
Collapse
Affiliation(s)
- Lindsay S Moore
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University, Stanford, CA, USA
| | - Esther de Boer
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Surgery, University of Groningen, Groningen, the Netherlands
| | - Andrew C Prince
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Neel Patel
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joshua M Richman
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anthony B Morlandt
- Department of Oral & Maxillofacial Surgery, University of Alabama Birmingham, Birmingham, AL, USA
| | - William R Carroll
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kurt R Zinn
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason M Warram
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA. .,Departments of Otolaryngology, Neurosurgery, and Radiology, The University of Alabama at Birmingham, 1670 University Blvd., Birmingham, AL, 35294, USA.
| |
Collapse
|
31
|
Mohapatra SS, Batra SK, Bharadwaj S, Bouvet M, Cosman B, Goel A, Jogunoori W, Kelley MJ, Mishra L, Mishra B, Mohapatra S, Patel B, Pisegna JR, Raufman JP, Rao S, Roy H, Scheuner M, Singh S, Vidyarthi G, White J. Precision Medicine for CRC Patients in the Veteran Population: State-of-the-Art, Challenges and Research Directions. Dig Dis Sci 2018; 63:1123-1138. [PMID: 29572615 PMCID: PMC5895694 DOI: 10.1007/s10620-018-5000-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/23/2018] [Indexed: 12/16/2022]
Abstract
Colorectal cancer (CRC) accounts for ~9% of all cancers in the Veteran population, a fact which has focused a great deal of the attention of the VA's research and development efforts. A field-based meeting of CRC experts was convened to discuss both challenges and opportunities in precision medicine for CRC. This group, designated as the VA Colorectal Cancer Cell-genomics Consortium (VA4C), discussed advances in CRC biology, biomarkers, and imaging for early detection and prevention. There was also a discussion of precision treatment involving fluorescence-guided surgery, targeted chemotherapies and immunotherapies, and personalized cancer treatment approaches. The overarching goal was to identify modalities that might ultimately lead to personalized cancer diagnosis and treatment. This review summarizes the findings of this VA field-based meeting, in which much of the current knowledge on CRC prescreening and treatment was discussed. It was concluded that there is a need and an opportunity to identify new targets for both the prevention of CRC and the development of effective therapies for advanced disease. Also, developing methods integrating genomic testing with tumoroid-based clinical drug response might lead to more accurate diagnosis and prognostication and more effective personalized treatment of CRC.
Collapse
Affiliation(s)
- Shyam S. Mohapatra
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- James A. Haley Veterans Hospital, Tampa, FL USA
- Division of Translational Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL USA
- College of Pharmacy Graduate Programs, University of South Florida, Tampa, FL USA
| | - Surinder K. Batra
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE USA
| | - Srinivas Bharadwaj
- Division of Translational Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| | - Michael Bouvet
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- VA San Diego Healthcare System, San Diego, CA USA
- Department of Surgery, University of California San Diego Moores Cancer Center, San Diego, CA USA
| | - Bard Cosman
- VA San Diego Healthcare System, San Diego, CA USA
- Department of Surgery, University of California San Diego Moores Cancer Center, San Diego, CA USA
| | - Ajay Goel
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute, Dallas, TX, USA
- Charles A. Sammons Cancer Center, Baylor University, Dallas, TX USA
| | - Wilma Jogunoori
- Washington DC VA Medical Center, Washington, DC USA
- Department of Surgery, Center for Translational Medicine, George Washington University, Washington, DC USA
| | - Michael J. Kelley
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- National Oncology Program Office, Specialty Care Services, Department of Veterans Affairs, Durham VA Medical Center, Durham, NC USA
- Department of Medicine, Duke University Medical Center, Durham, NC USA
| | - Lopa Mishra
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- Washington DC VA Medical Center, Washington, DC USA
- Department of Surgery, Center for Translational Medicine, George Washington University, Washington, DC USA
| | - Bibhuti Mishra
- Washington DC VA Medical Center, Washington, DC USA
- Department of Surgery, Center for Translational Medicine, George Washington University, Washington, DC USA
| | - Subhra Mohapatra
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- James A. Haley Veterans Hospital, Tampa, FL USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| | - Bhaumik Patel
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- Hunter Holmes McGuire VA Medical Center and Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA USA
| | - Joseph R. Pisegna
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- Division of Gastroenterology and Human Genetics, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Jean-Pierre Raufman
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- VA Maryland Health Care System, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Shuyun Rao
- Washington DC VA Medical Center, Washington, DC USA
- Department of Surgery, Center for Translational Medicine, George Washington University, Washington, DC USA
| | - Hemant Roy
- Department of Medicine, Boston University School of Medicine, Boston, MA USA
| | - Maren Scheuner
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- Division of Gastroenterology and Human Genetics, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Satish Singh
- Department of Veterans Affairs Colorectal Cancer Cell-genomics Consortium [VA4C], Tampa, FL USA
- VA Boston Healthcare System and Department of Medicine, Boston University School of Medicine, Boston, MA USA
| | - Gitanjali Vidyarthi
- James A. Haley Veterans Hospital, Tampa, FL USA
- Division of Translational Medicine, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| | - Jon White
- Washington DC VA Medical Center, Washington, DC USA
- Department of Surgery, Center for Translational Medicine, George Washington University, Washington, DC USA
| |
Collapse
|
32
|
Expression and Targeting of Tumor Markers in Gelfoam ® Histoculture: Potential Individualized Assays for Immuno-Oncology. Methods Mol Biol 2018. [PMID: 29572791 DOI: 10.1007/978-1-4939-7745-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Tumor-specific antigens are important in the study of tumor biology, tumor diagnosis, and prognosis and as targets for tumor therapy. This chapter reviews patient colon, breast, and ovarian tumors in 3-dimensional Gelfoam® histoculture maintaining in vivo-like expression of the important tumor antigens, for example TAG-72 and CEA. We have also reviewed that fluorescent antibodies can target tumors in Gelfoam® histoculture, thereby providing an assay for individual patients for sensitivity to therapeutic antibodies which have become so important in immuno-oncology and other cancer therapies.
Collapse
|
33
|
Sun Y, Zeng X, Xiao Y, Liu C, Zhu H, Zhou H, Chen Z, Xu F, Wang J, Zhu M, Wu J, Tian M, Zhang H, Deng Z, Cheng Z, Hong X. Novel dual-function near-infrared II fluorescence and PET probe for tumor delineation and image-guided surgery. Chem Sci 2018; 9:2092-2097. [PMID: 29675250 PMCID: PMC5892408 DOI: 10.1039/c7sc04774f] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 12/27/2017] [Indexed: 12/25/2022] Open
Abstract
The first small-molecule based αvβ3-targeted NIR-II/PET dual-modal probes via base-catalyzed thiol-addition chemistry were concisely assembled and evaluated.
Accurate tumor identification is essential in cancer management. Incomplete excision of tumor tissue, however, negatively affects the prognosis of the patient. To accomplish radical excision of tumor tissue, radiotracers can be used that target tumor tissue and can be detected using a gamma probe during surgery. Intraoperative fluorescence imaging could allow accurate real-time tumor delineation. Herein, a novel dual-modal imaging platform using base-catalyzed double addition of thiols into a propiolamide scaffold has been developed, allowing for the highly efficient and selective assembly of various thiol units in a protecting-group-free manner. The first small-molecule based αvβ3-targeted NIR-II/PET probe 68Ga-SCH2 was concisely generated via this strategy and subsequently evaluated in mice bearing the U87MG xenograft. Excellent imaging properties such as good tumor uptake, high tumor contrast and specificity, tumor delineation and image-guided surgery were achieved in the small animal models. These attractive results of 68Ga-SCH2 allow it to be a promising αvβ3-targeted NIR-II/PET probe for clinical translation.
Collapse
Affiliation(s)
- Yao Sun
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China . .,Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , College of Chemistry , Central China Normal University , Wuhan 430079 , China
| | - Xiaodong Zeng
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Yuling Xiao
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Changhao Liu
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Stanford University , CA 94305 , USA .
| | - Hua Zhu
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Stanford University , CA 94305 , USA .
| | - Hui Zhou
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Ziyang Chen
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Fuchun Xu
- Medical College , Tibet University , Lasa , 850000 , China
| | - Jule Wang
- Medical College , Tibet University , Lasa , 850000 , China
| | - Mengyue Zhu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease , Center for Experimental Basic Medical Education , Wuhan University , Wuhan 430071 , China
| | - Junzhu Wu
- Hubei Provincial Key Laboratory of Developmentally Originated Disease , Center for Experimental Basic Medical Education , Wuhan University , Wuhan 430071 , China
| | - Mei Tian
- Department of Nuclear Medicine , The Second Hospital of Zhejiang University School of Medicine , 88 Jiefang Road , Hangzhou , 310009 , China .
| | - Hong Zhang
- Department of Nuclear Medicine , The Second Hospital of Zhejiang University School of Medicine , 88 Jiefang Road , Hangzhou , 310009 , China .
| | - Zixin Deng
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Stanford University , CA 94305 , USA .
| | - Xuechuan Hong
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China . .,Medical College , Tibet University , Lasa , 850000 , China
| |
Collapse
|
34
|
DeLong JC, Murakami T, Yazaki PJ, Hoffman RM, Bouvet M. Near-infrared-conjugated humanized anti-carcinoembryonic antigen antibody targets colon cancer in an orthotopic nude-mouse model. J Surg Res 2017; 218:139-143. [PMID: 28985840 DOI: 10.1016/j.jss.2017.05.069] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 04/27/2017] [Accepted: 05/19/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND The success of a curative surgery for cancer is dependent on the complete removal of all cancer cells. Tumor visualization by the surgeon can be enhanced through fluorescent-antibody targeting. To further develop such technology, we selected humanized anti-carcinoembryonic antigen (CEA) conjugated to a near-infrared dye to target orthotopically-implanted human colon cancer in nude mice. MATERIALS AND METHODS The HT-29 human colon cancer cell line was grown in culture and subcutaneously injected in mice. After 3 wk of growth, tumors were resected and cut into 2 mm3 fragments that were sutured to the cecum of five additional nude mice for orthotopic implantation. The tumors were allowed to grow for 4 wk at which point 3 had successful orthotopic tumor growth and were selected for injection of the humanized anti-CEA antibody conjugated to the near-infrared dye IRDye800CW (anti-CEA-IRDye800CW). The antibody-dye conjugate (75 μg) was administered via tail vein injection. Images were obtained with the Pearl Trilogy Small Animal Imaging System with both 700 and 800 nm channels and evaluated using Image Studio. RESULTS Laparotomy was performed 24 h after labeling the tumors. When imaged through the 800 nm channel, the tumors were observed to be strongly labeled with anti-CEA-IRDye800. At 48 h, laparotomy was repeated which again demonstrated strong labeling of the tumors through the 800 nm channel, but with a lower absolute intensity (in relative units), than at 24 h. CONCLUSIONS Humanized anti-CEA-IRDye800CW can rapidly and effectively label CEA-expressing human colon cancer in an orthotopic nude mouse model. Given the ability of this technology to target and label tumors with great specificity, the anti-CEA-IRDye800CW is currently being developed for clinical use in fluorescence-guided surgery.
Collapse
Affiliation(s)
- Jonathan C DeLong
- Department of Surgery, University of California San Diego, San Diego, California
| | - Takashi Murakami
- Department of Surgery, University of California San Diego, San Diego, California; AntiCancer, Inc, San Diego, California; Department of Surgery, Yokohama City University, Yokohama City, Japan
| | - Paul J Yazaki
- Beckman Research Institute, City of Hope Medical Center, Duarte, California
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, San Diego, California; AntiCancer, Inc, San Diego, California
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, California; Department of Surgery, VA Healthcare System, San Diego, California.
| |
Collapse
|
35
|
Sheng W, He S, Seare WJ, Almutairi A. Review of the progress toward achieving heat confinement-the holy grail of photothermal therapy. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:80901. [PMID: 28776627 PMCID: PMC5544355 DOI: 10.1117/1.jbo.22.8.080901] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/23/2017] [Indexed: 06/01/2023]
Abstract
Photothermal therapy (PTT) involves the application of normally benign light wavelengths in combination with efficient photothermal (PT) agents that convert the absorbed light to heat to ablate selected cancers. The major challenge in PTT is the ability to confine heating and thus direct cellular death to precisely where PT agents are located. The dominant strategy in the field has been to create large libraries of PT agents with increased absorption capabilities and to enhance their delivery and accumulation to achieve sufficiently high concentrations in the tissue targets of interest. While the challenge of material confinement is important for achieving “heat and lethality confinement,” this review article suggests another key prospective strategy to make this goal a reality. In this approach, equal emphasis is placed on selecting parameters of light exposure, including wavelength, duration, power density, and total power supplied, based on the intrinsic properties and geometry of tissue targets that influence heat dissipation, to truly achieve heat confinement. This review highlights significant milestones researchers have achieved, as well as examples that suggest future research directions, in this promising technique, as it becomes more relevant in clinical cancer therapy and other noncancer applications.
Collapse
Affiliation(s)
- Wangzhong Sheng
- University of California, Laboratory for Bioresponsive Materials, Department of Mechanical and Aerospace Engineering, Materials Science Program, La Jolla, San Diego, California, United States
- University of California, Laboratory for Bioresponsive Materials, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, San Diego, California, United States
| | - Sha He
- University of California, Laboratory for Bioresponsive Materials, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, San Diego, California, United States
- University of California, Laboratory for Bioresponsive Materials, Department of Nanoengineering, La Jolla, San Diego, California, United States
| | | | - Adah Almutairi
- University of California, Laboratory for Bioresponsive Materials, Department of Mechanical and Aerospace Engineering, Materials Science Program, La Jolla, San Diego, California, United States
- University of California, Laboratory for Bioresponsive Materials, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, San Diego, California, United States
- University of California, Laboratory for Bioresponsive Materials, Department of Nanoengineering, La Jolla, San Diego, California, United States
| |
Collapse
|
36
|
SGM-101: An innovative near-infrared dye-antibody conjugate that targets CEA for fluorescence-guided surgery. Surg Oncol 2017; 26:153-162. [DOI: 10.1016/j.suronc.2017.03.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/06/2017] [Accepted: 03/06/2017] [Indexed: 12/31/2022]
|
37
|
Sun Y, Ding M, Zeng X, Xiao Y, Wu H, Zhou H, Ding B, Qu C, Hou W, Er-Bu A, Zhang Y, Cheng Z, Hong X. Novel bright-emission small-molecule NIR-II fluorophores for in vivo tumor imaging and image-guided surgery. Chem Sci 2017; 8:3489-3493. [PMID: 28507722 PMCID: PMC5418643 DOI: 10.1039/c7sc00251c] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 02/18/2017] [Indexed: 12/22/2022] Open
Abstract
Though high brightness and biocompatible small NIR-II dyes are highly desirable in clinical or translational cancer research, their fluorescent cores are relatively limited and their synthetic processes are somewhat complicated. Herein, we have explored the design and synthesis of novel NIR-II fluorescent materials (H1) without tedious chromatographic isolation with improved fluorescence performance (QY ≈ 2%) by introducing 2-amino 9,9-dialkyl-substituted fluorene as a donor into the backbone. Several types of water-soluble and biocompatible NIR-II probes: SXH, SDH, and H1 NPs were constructed via different chemical strategies based on H1, and then their potential to be used in in vivo tumor imaging and image-guided surgery in the NIR-II region was explored. High levels of uptake were obtained for both passive and active tumor targeting probes SXH and SDH. Furthermore, high resolution imaging of blood vessels on tumors and the whole body of living mice using H1 NPs for the first time has demonstrated precise NIR-II image-guided sentinel lymph node (SLN) surgery.
Collapse
Affiliation(s)
- Yao Sun
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
- Key Laboratory of Pesticides and Chemical Biology , Ministry of Education , College of Chemistry , Central China Normal University , Wuhan 430079 , China
| | - Mingmin Ding
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Xiaodong Zeng
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Yuling Xiao
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Huaping Wu
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Hui Zhou
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Bingbing Ding
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Chunrong Qu
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Wei Hou
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Aga Er-Bu
- Medical College , Tibet University , Lasa , 850000 , China
| | - Yejun Zhang
- Suzhou NIR-Optics Technologies Co., Ltd , Suzhou , 215123 , China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS) , Bio-X Program , Department of Radiology , Stanford University , California 94305-5344 , USA
| | - Xuechuan Hong
- State Key Laboratory of Virology , Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) , Hubei Provincial Key Laboratory of Developmentally Originated Disease , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
- Medical College , Tibet University , Lasa , 850000 , China
| |
Collapse
|
38
|
Park JY, Lee JY, Zhang Y, Hoffman RM, Bouvet M. Targeting the insulin growth factor-1 receptor with fluorescent antibodies enables high resolution imaging of human pancreatic cancer in orthotopic mouse models. Oncotarget 2017; 7:18262-8. [PMID: 26919100 PMCID: PMC4951286 DOI: 10.18632/oncotarget.7576] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/11/2016] [Indexed: 12/11/2022] Open
Abstract
The goal of the present study was to determine whether insulin-like growth factor-1 receptor (IGF-1R) antibodies, conjugated with bright fluorophores, could enable visualization of pancreatic cancer in orthotopic nude mouse models. IGF-1R antibody (clone 24-31) was conjugated with 550 nm or 650 nm fluorophores. Western blotting confirmed the expression of IGF-1R in Panc-1, BxPC3, and MIAPaCa-2 human pancreatic cancer cell lines. Labeling with fluorophore-conjugated IGF-1R antibody demonstrated fluorescent foci on the membrane of the pancreatic cancer cells. Subcutaneous Panc-1, BxPC-3, and MIA PaCa-2 tumors became fluorescent after intravenous administration of fluorescent IGF-1R antibodies. Orthotopically-transplanted BxPC-3 tumors became fluorescent with the conjugated IGF-1R antibodies, and were easily visible with intravital imaging. Gross and microscopic ex vivo imaging of resected pancreatic tumor and normal pancreas confirmed that fluorescence indeed came from the membrane of cancer cells, and it was stronger from the tumor than the normal tissue. The present study demonstrates that fluorophore-conjugated IGF-1R antibodies can visualize pancreatic cancer and it can be used with various imaging devices such as endoscopy and laparoscopy for diagnosis and fluorescence-guided surgery.
Collapse
Affiliation(s)
- Jeong Youp Park
- Department of Surgery, University of California San Diego, San Diego, CA, USA.,AntiCancer, Inc., San Diego, CA, USA.,Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Young Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | | | - Robert M Hoffman
- Department of Surgery, University of California San Diego, San Diego, CA, USA.,AntiCancer, Inc., San Diego, CA, USA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, CA, USA.,Surgical Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
39
|
Keating JJ, Okusanya OT, De Jesus E, Judy R, Jiang J, Deshpande C, Nie S, Low P, Singhal S. Intraoperative Molecular Imaging of Lung Adenocarcinoma Can Identify Residual Tumor Cells at the Surgical Margins. Mol Imaging Biol 2016; 18:209-18. [PMID: 26228697 DOI: 10.1007/s11307-015-0878-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE During lung surgery, identification of surgical margins is challenging. We hypothesized that molecular imaging with a fluorescent probe to pulmonary adenocarcinomas could enhance residual tumor during resection. PROCEDURES Mice with flank tumors received a contrast agent targeting folate receptor alpha. Optimal dose and time of injection was established. Margin detection was compared using traditional methods versus molecular imaging. A pilot study was then performed in three humans with lung adenocarcinoma. RESULTS The peak tumor-to-background ratio (TBR) of murine tumors was 3.9. Fluorescence peaked at 2 h and was not improved beyond 0.1 mg/kg. Traditional inspection identified 30% of mice with positive margins. Molecular imaging identified an additional 50% of residual tumor deposits (p < 0.05). The fluorescent probe visually enhanced all human tumors with a mean TBR of 3.5. CONCLUSIONS Molecular imaging is an important adjunct to traditional inspection to identify surgical margins after tumor resection.
Collapse
Affiliation(s)
- Jane J Keating
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania and Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Olugbenga T Okusanya
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania and Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Elizabeth De Jesus
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania and Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Ryan Judy
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania and Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Jack Jiang
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania and Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Charuhas Deshpande
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuming Nie
- Departments of Biomedical Engineering and Chemistry, Emory University, Atlanta, GA, USA
| | - Philip Low
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania and Philadelphia VA Medical Center, Philadelphia, PA, USA.
- Division of Thoracic Surgery, University of Pennsylvania School of Medicine, 6 White Building, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
40
|
Diana M, Robinet E, Liu YY, Legnèr A, Kong SH, Schiraldi L, Marchegiani F, Halvax P, Swanstrom L, Dallemagne B, Marescaux J. Confocal Imaging and Tissue-Specific Fluorescent Probes for Real-Time In Vivo Immunohistochemistry. Proof of the Concept in a Gastric Lymph Node Metastasis Model. Ann Surg Oncol 2016; 23:567-573. [PMID: 26511264 DOI: 10.1245/s10434-015-4928-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Indexed: 04/13/2025]
Abstract
BACKGROUND Tumor-specific fluorescent antibodies, which can be recognized at a cellular or tissue level using optical imaging such as confocal laser endomicroscopy (CLE), could provide a means for rapid and accurate tumor diagnosis and staging. The aim of this study was to evaluate the ability of CLE to detect the presence of tagged cells within lymph nodes in an original simulated metastatic model. MATERIALS AND METHODS A solution of indocyanine green containing a suspension of porcine hepatocytes, marked with carboxy-fluorescein-succinimidyl-ester (CFSE), was injected endoscopically in the gastric submucosa of 10 pigs. Fluorescence lymphography using a near-infrared laparoscope was used to identify sentinel and secondary drainage nodes. Additionally, a nonfluorescent gastric and a mesenteric node were identified. Every 5-10 min, those nodes were scanned using probe-based or needle-based CLE (pCLE or nCLE). Immunohistochemistry (IHC) using anti-cytokeratin 18 antibodies was subsequently performed to confirm the presence of hepatocytes in the lymph nodes. RESULTS A total of 36 lymph nodes were analyzed with both CLE probes. Hepatocyte penetration in lymph nodes, as assessed by repeated CLE scanning, took 10-40 min after submucosal injection. Concordance between CLE and IHC was 84 and 72 % for pCLE and nCLE, respectively. False negatives were partly due to incomplete CFSE labeling of hepatocytes, which could not be recognized by CLE, but were detected with IHC. CONCLUSIONS Real-time CLE analysis effectively recognized the presence in perigastric nodes of marked hepatic cells that had been injected endoscopically in the stomach. Validation studies on tumor-bearing animals using tumor-specific antibodies should be performed.
Collapse
Affiliation(s)
- Michele Diana
- Research Institute Against Cancer of the Digestive System, IRCAD, Strasbourg, France.
- Institute of Image-Guided Surgery, IHU-Strasbourg, Strasbourg, France.
| | - Eric Robinet
- Institute of Image-Guided Surgery, IHU-Strasbourg, Strasbourg, France
| | - Yu-Yin Liu
- Research Institute Against Cancer of the Digestive System, IRCAD, Strasbourg, France
- Department of General Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Andras Legnèr
- Institute of Image-Guided Surgery, IHU-Strasbourg, Strasbourg, France
| | - Seong-Ho Kong
- Institute of Image-Guided Surgery, IHU-Strasbourg, Strasbourg, France
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Luigi Schiraldi
- Research Institute Against Cancer of the Digestive System, IRCAD, Strasbourg, France
| | - Francesco Marchegiani
- Research Institute Against Cancer of the Digestive System, IRCAD, Strasbourg, France
| | - Peter Halvax
- Institute of Image-Guided Surgery, IHU-Strasbourg, Strasbourg, France
| | - Lee Swanstrom
- Institute of Image-Guided Surgery, IHU-Strasbourg, Strasbourg, France
| | - Bernard Dallemagne
- Research Institute Against Cancer of the Digestive System, IRCAD, Strasbourg, France
| | - Jacques Marescaux
- Research Institute Against Cancer of the Digestive System, IRCAD, Strasbourg, France
- Institute of Image-Guided Surgery, IHU-Strasbourg, Strasbourg, France
| |
Collapse
|
41
|
de Geus SWL, Boogerd LSF, Swijnenburg RJ, Mieog JSD, Tummers WSFJ, Prevoo HAJM, Sier CFM, Morreau H, Bonsing BA, van de Velde CJH, Vahrmeijer AL, Kuppen PJK. Selecting Tumor-Specific Molecular Targets in Pancreatic Adenocarcinoma: Paving the Way for Image-Guided Pancreatic Surgery. Mol Imaging Biol 2016; 18:807-819. [PMID: 27130234 PMCID: PMC5093212 DOI: 10.1007/s11307-016-0959-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE The purpose of this study was to identify suitable molecular targets for tumor-specific imaging of pancreatic adenocarcinoma. PROCEDURES The expression of eight potential imaging targets was assessed by the target selection criteria (TASC)-score and immunohistochemical analysis in normal pancreatic tissue (n = 9), pancreatic (n = 137), and periampullary (n = 28) adenocarcinoma. RESULTS Integrin αvβ6, carcinoembryonic antigen (CEA), epithelial growth factor receptor (EGFR), and urokinase plasminogen activator receptor (uPAR) showed a significantly higher (all p < 0.001) expression in pancreatic adenocarcinoma compared to normal pancreatic tissue and were confirmed by the TASC score as promising imaging targets. Furthermore, these biomarkers were expressed in respectively 88 %, 71 %, 69 %, and 67 % of the pancreatic adenocarcinoma patients. CONCLUSIONS The results of this study show that integrin αvβ6, CEA, EGFR, and uPAR are suitable targets for tumor-specific imaging of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Susanna W L de Geus
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Leonora S F Boogerd
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Rutger-Jan Swijnenburg
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - J Sven D Mieog
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Willemieke S F J Tummers
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Hendrica A J M Prevoo
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Cornelis F M Sier
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Cornelis J H van de Velde
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Alexander L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
42
|
Hiroshima Y, Lwin TM, Murakami T, Mawy AA, Kuniya T, Chishima T, Endo I, Clary BM, Hoffman RM, Bouvet M. Effective fluorescence-guided surgery of liver metastasis using a fluorescent anti-CEA antibody. J Surg Oncol 2016; 114:951-958. [PMID: 27696448 PMCID: PMC5565879 DOI: 10.1002/jso.24462] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 09/11/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND OBJECTIVES Delineation of adequate tumor margins is critical in oncologic surgery, particularly in resection of metastatic lesions. Surgeons are limited in visualization with bright-light surgery, but fluorescence-guided surgery (FGS) has been efficacious in helping the surgeon achieve negative margins. METHODS The present study uses FGS in a mouse model that has undergone surgical orthotopic implantation (SOI) of colorectal liver metastasis tagged with green fluorescent protein (GFP). An anti-CEA antibody conjugated to DyLight 650 was used to highlight the tumor. RESULTS The fluorescent antibody clearly demarcated the lesion at deeper tissue depth compared to GFP. Fluorescence of the anti-CEA-DyLight650 showed maximal tumor-to-liver contrast at 72 hr. Fifteen mice underwent bright-light surgery (BLS) versus FGS with GFP versus FGS with anti-CEA-DyLight650. Mice that underwent FGS had a significantly smaller area of residual tumor (P < 0.001) and significantly longer overall survival (P < 0.001) and disease-free survival (P < 0.001). Within the two FGS groups, mice undergoing surgery with anti-CEA-DyLight650 improved survival compared to only GFP labeling. CONCLUSIONS In the present report, we demonstrate that an anti-CEA antibody conjugated to a DyLight 650 nm dye clearly labeled colon cancer liver metastases, thereby enabling successful FGS. J. Surg. Oncol. 2016;114:951-958. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yukihiko Hiroshima
- Department of Surgery, University of California San Diego, San Diego, California
- AntiCancer, Inc., San Diego, California
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Thinzar M. Lwin
- Department of Surgery, University of California San Diego, San Diego, California
- AntiCancer, Inc., San Diego, California
| | - Takashi Murakami
- Department of Surgery, University of California San Diego, San Diego, California
- AntiCancer, Inc., San Diego, California
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Ali A. Mawy
- Department of Surgery, University of California San Diego, San Diego, California
- AntiCancer, Inc., San Diego, California
| | - Tanaka Kuniya
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Takashi Chishima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Bryan M. Clary
- Department of Surgery, University of California San Diego, San Diego, California
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, San Diego, California
- AntiCancer, Inc., San Diego, California
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, California
| |
Collapse
|
43
|
Zhang YF, Ho M. Humanization of high-affinity antibodies targeting glypican-3 in hepatocellular carcinoma. Sci Rep 2016; 6:33878. [PMID: 27667400 PMCID: PMC5036187 DOI: 10.1038/srep33878] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/05/2016] [Indexed: 01/03/2023] Open
Abstract
Glypican-3 (GPC3) is a cell-surface heparan sulfate proteoglycan highly expressed in hepatocellular carcinoma (HCC). We have generated a group of high-affinity mouse monoclonal antibodies targeting GPC3. Here, we report the humanization and testing of these antibodies for clinical development. We compared the affinity and cytotoxicity of recombinant immunotoxins containing mouse single-chain variable regions fused with a Pseudomonas toxin. To humanize the mouse Fvs, we grafted the combined KABAT/IMGT complementarity determining regions (CDR) into a human IgG germline framework. Interestingly, we found that the proline at position 41, a non-CDR residue in heavy chain variable regions (VH), is important for humanization of mouse antibodies. We also showed that two humanized anti-GPC3 antibodies (hYP7 and hYP9.1b) in the IgG format induced antibody-dependent cell-mediated cytotoxicity and complement-dependent-cytotoxicity in GPC3-positive cancer cells. The hYP7 antibody was tested and showed inhibition of HCC xenograft tumor growth in nude mice. This study successfully humanizes and validates high affinity anti-GPC3 antibodies and sets a foundation for future development of these antibodies in various clinical formats in the treatment of liver cancer.
Collapse
Affiliation(s)
- Yi-Fan Zhang
- Antibody Therapy Section, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Mitchell Ho
- Antibody Therapy Section, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
44
|
Saccomano M, Dullin C, Alves F, Napp J. Preclinical evaluation of near-infrared (NIR) fluorescently labeled cetuximab as a potential tool for fluorescence-guided surgery. Int J Cancer 2016; 139:2277-89. [PMID: 27428782 DOI: 10.1002/ijc.30277] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/01/2016] [Accepted: 06/23/2016] [Indexed: 02/06/2023]
Abstract
The high rate of recurrence in patients with pancreatic ductal adenocarcinoma (PDAC) could be reduced by supporting the surgeons in discriminating healthy from diseased tissues with intraoperative fluorescence-guidance. Here, we studied the suitability of Cetuximab, a therapeutic monoclonal antibody targeting the human epidermal growth factor receptor (EGFR), near-infrared (NIR) fluorescently labeled as a new tool for fluorescence-guided surgery. Distribution and binding of systemically injected Cetuximab Alexa Fluor 647 conjugate (Cetux-Alexa-647) and the co-injected control human IgG Alexa Fluor 750 conjugate (hIgG-Alexa-750) was studied over 48 h by NIR fluorescence imaging in mice bearing human orthotopic AsPC-1 and MIA PaCa-2 PDAC tumors. Cetux-Alexa-647, but not the control hIgG-Alexa-750 fluorescence, was specifically detected in vivo in both primary pancreatic tumors with maximum fluorescence intensities at 24 h, and in metastases of AsPC-1 tumors as small as 1 mm. Lifetime analysis and NIR fluorescence microscopy of tumor sections confirmed the binding specificity of Cetux-Alexa-647 to PDAC cells. Comparable results were obtained with Cetuximab conjugated to Alexa Fluor 750 dye (Cetux-Alexa-750). Fluorescence-guided dissection, performed 24 h after injection of Cetuximab conjugated to IRDye 800CW (Cetux-800CW), enabled a real-time delineation of AsPC-1 tumor margins, and small metastases. Odyssey scans revealed that only the vital part of the tumor, but not the necrotic part was stained with Cetux-800CW. NIR fluorescently labeled Cetuximab may be a promising tool that can be applied for fluorescence-guided surgery to visualize tumor margins and metastatic sites in order to allow a precise surgical resection.
Collapse
Affiliation(s)
- Mara Saccomano
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Christian Dullin
- Institute of Interventional and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Frauke Alves
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany.,Institute of Interventional and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany.,Department of Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Joanna Napp
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany. .,Institute of Interventional and Diagnostic Radiology, University Medical Center Göttingen, Göttingen, Germany. .,Department of Haematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
45
|
Knutson S, Raja E, Bomgarden R, Nlend M, Chen A, Kalyanasundaram R, Desai S. Development and Evaluation of a Fluorescent Antibody-Drug Conjugate for Molecular Imaging and Targeted Therapy of Pancreatic Cancer. PLoS One 2016; 11:e0157762. [PMID: 27336622 PMCID: PMC4918962 DOI: 10.1371/journal.pone.0157762] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/03/2016] [Indexed: 11/18/2022] Open
Abstract
Antibodies are widely available and cost-effective research tools in life science, and antibody conjugates are now extensively used for targeted therapy, immunohistochemical staining, or in vivo diagnostic imaging of cancer. Significant advances in site-specific antibody labeling technologies have enabled the production of highly characterized and homogenous conjugates for biomedical purposes, and some recent studies have utilized site-specific labeling to synthesize bifunctional antibody conjugates with both imaging and drug delivery properties. While these advances are important for the clinical safety and efficacy of such biologics, these techniques can also be difficult, expensive, and time-consuming. Furthermore, antibody-drug conjugates (ADCs) used for tumor treatment generally remain distinct from conjugates used for diagnosis. Thus, there exists a need to develop simple dual-labeling methods for efficient therapeutic and diagnostic evaluation of antibody conjugates in pre-clinical model systems. Here, we present a rapid and simple method utilizing commercially available reagents for synthesizing a dual-labeled fluorescent ADC. Further, we demonstrate the fluorescent ADC’s utility for simultaneous targeted therapy and molecular imaging of cancer both in vitro and in vivo. Employing non-site-specific, amine-reactive chemistry, our novel biopharmaceutical theranostic is a monoclonal antibody specific for a carcinoembryonic antigen (CEA) biomarker conjugated to both paclitaxel and a near-infrared (NIR), polyethylene glycol modified (PEGylated) fluorophore (DyLight™ 680-4xPEG). Using in vitro systems, we demonstrate that this fluorescent ADC selectively binds a CEA-positive pancreatic cancer cell line (BxPC-3) in immunofluorescent staining and flow cytometry, exhibits efficient internalization kinetics, and is cytotoxic. Model studies using a xenograft of BxPC-3 cells in athymic mice also show the fluorescent ADC’s efficacy in detecting tumors in vivo and inhibiting tumor growth more effectively than equimolar amounts of unconjugated drug. Overall, our results demonstrate that non-selective, amine-targeting chemistry is an effective dual-labeling method for synthesizing and evaluating a bifunctional fluorescent antibody-drug conjugate, allowing concurrent detection, monitoring and treatment of cancer.
Collapse
Affiliation(s)
- Steve Knutson
- Department of Research and Development, Thermo Fisher Scientific, Rockford, Illinois, United States of America
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, United States of America
- * E-mail: (SK); (SD)
| | - Erum Raja
- Department of Research and Development, Thermo Fisher Scientific, Rockford, Illinois, United States of America
| | - Ryan Bomgarden
- Department of Research and Development, Thermo Fisher Scientific, Rockford, Illinois, United States of America
| | - Marie Nlend
- Department of Research and Development, Thermo Fisher Scientific, Rockford, Illinois, United States of America
| | - Aoshuang Chen
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, United States of America
| | - Ramaswamy Kalyanasundaram
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, United States of America
| | - Surbhi Desai
- Department of Research and Development, Thermo Fisher Scientific, Rockford, Illinois, United States of America
- * E-mail: (SK); (SD)
| |
Collapse
|
46
|
Abstract
The surgical management of pancreatic diseases is rapidly evolving, encompassing advances in evidence-driven selection of patients amenable for surgical therapy, preoperative risk stratification, refinements in the technical conduct of pancreatic operations, and quantification of postoperative morbidity. These advances have resulted in dramatic reductions in mortality following pancreatic surgery, particularly at high-volume pancreatic centers. Surgical decision making is complex, and requires an intimate understanding of disease pathobiology, host physiology, technical considerations, and evolving trends. This article highlights key developments in the contemporary surgical management of pancreatic diseases.
Collapse
Affiliation(s)
- Jashodeep Datta
- Division of Gastrointestinal Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Charles M Vollmer
- Division of Gastrointestinal Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
47
|
Park JY, Murakami T, Lee JY, Zhang Y, Hoffman RM, Bouvet M. Fluorescent-Antibody Targeting of Insulin-Like Growth Factor-1 Receptor Visualizes Metastatic Human Colon Cancer in Orthotopic Mouse Models. PLoS One 2016; 11:e0146504. [PMID: 26731105 PMCID: PMC4701661 DOI: 10.1371/journal.pone.0146504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 12/17/2015] [Indexed: 12/19/2022] Open
Abstract
Fluorescent-antibody targeting of metastatic cancer has been demonstrated by our laboratory to enable tumor visualization and effective fluorescence-guided surgery. The goal of the present study was to determine whether insulin-like growth factor-1 receptor (IGF-1R) antibodies, conjugated with bright fluorophores, could enable visualization of metastatic colon cancer in orthotopic nude mouse models. IGF-1R antibody (clone 24–31) was conjugated with 550 nm, 650 nm or PEGylated 650 nm fluorophores. Subcutaneous, orthotopic, and liver metastasis models of colon cancer in nude mice were targeted with the fluorescent IGF-1R antibodies. Western blotting confirmed the expression of IGF-1R in HT-29 and HCT 116 human colon cancer cell lines, both expressing green fluorescent protein (GFP). Labeling with fluorophore-conjugated IGF-1R antibody demonstrated fluorescent foci on the membrane of colon cancer cells. Subcutaneously- and orthotopically-transplanted HT-29-GFP and HCT 116-GFP tumors brightly fluoresced at the longer wavelengths after intravenous administration of fluorescent IGF-1R antibodies. Orthotopically-transplanted HCT 116-GFP tumors were brightly labeled by fluorescent IGF-1R antibodies such that they could be imaged non-invasively at the longer wavelengths. In an experimental liver metastasis model, IGF-1R antibodies conjugated with PEGylated 650 nm fluorophores selectively highlighted the liver metastases, which could then be non-invasively imaged. The IGF-1R fluorescent-antibody labeled liver metastases were very bright compared to the normal liver and the fluorescent-antibody label co-located with green fluorescent protein (GFP) expression of the colon cancer cells. The present study thus demonstrates that fluorophore-conjugated IGF-1R antibodies selectively visualize metastatic colon cancer and have clinical potential for improved diagnosis and fluorescence-guided surgery.
Collapse
Affiliation(s)
- Jeong Youp Park
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Takashi Murakami
- AntiCancer, Inc., San Diego, California, United States of America
- Department of Surgery, Yokohama City University Graduate School of Medicine, Yokohama City, Japan
| | - Jin Young Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Zhang
- AntiCancer, Inc., San Diego, California, United States of America
| | - Robert M. Hoffman
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
- AntiCancer, Inc., San Diego, California, United States of America
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, California, United States of America
- Surgical Service, VA San Diego Healthcare System, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
48
|
DeLong JC, Hoffman RM, Bouvet M. Current status and future perspectives of fluorescence-guided surgery for cancer. Expert Rev Anticancer Ther 2015; 16:71-81. [PMID: 26567611 DOI: 10.1586/14737140.2016.1121109] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Curative cancer surgery is dependent on the removal of all primary tumor and metastatic cancer cells. Preoperative imaging, intraoperative inspection and palpation, as well as pathological margin confirmation aid the surgeon, but these methods are lacking in sensitivity and can be highly subjective. Techniques in fluorescence-guided surgery (FGS) are emerging that selectively illuminate cancer cells, enhancing the distinction between tumors and surrounding tissues with the potential for single-cell sensitivity. FGS enhances tumor detection, surgical navigation, margin confirmation, and in some cases can be combined with therapeutic techniques to eliminate microscopic disease. In this review, we describe the preclinical developments and currently-used techniques for FGS.
Collapse
Affiliation(s)
- Jonathan C DeLong
- a Department of Surgery , University of California San Diego , San Diego , CA , USA
| | - Robert M Hoffman
- a Department of Surgery , University of California San Diego , San Diego , CA , USA.,b AntiCancer, Inc ., San Diego , CA , USA
| | - Michael Bouvet
- a Department of Surgery , University of California San Diego , San Diego , CA , USA
| |
Collapse
|
49
|
Yano S, Zhang Y, Miwa S, Kishimoto H, Urata Y, Bouvet M, Kagawa S, Fujiwara T, Hoffman RM. Precise navigation surgery of tumours in the lung in mouse models enabled by in situ fluorescence labelling with a killer-reporter adenovirus. BMJ Open Respir Res 2015; 2:e000096. [PMID: 26380093 PMCID: PMC4567685 DOI: 10.1136/bmjresp-2015-000096] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 07/08/2015] [Accepted: 07/10/2015] [Indexed: 01/18/2023] Open
Abstract
Background Current methods of image-guided surgery of tumours of the lung mostly rely on CT. A sensitive procedure of selective tumour fluorescence labelling would allow simple and high-resolution visualisation of the tumour for precise surgical navigation. Methods Human lung cancer cell lines H460 and A549 were genetically transformed to express red fluorescent protein (RFP). Tumours were grown subcutaneously for each cell line and harvested and minced for surgical orthotopic implantation on the left lung of nude mice. Tumour growth was measured by fluorescence imaging. After the tumours reached 5 mm in diameter, they were injected under fluorescence guidance with the telomerase-dependent green fluorescent protein (GFP)-containing adenovirus, OBP-401. Viral labelling of the lung tumours with GFP precisely colocalised with tumour RFP expression. Three days after administration of OBP-401, fluorescence-guided surgery (FGS) was performed. Results FGS of tumours in the lung was enabled by labelling with a telomerase-dependent adenovirus containing the GFP gene. Tumours in the lung were selectively and brightly labelled. FGS enabled complete lung tumour resection with no residual fluorescent tumour. Conclusions FGS of tumours in the lung is feasible and more effective than bright-light surgery.
Collapse
Affiliation(s)
- Shuya Yano
- AntiCancer, Inc. , San Diego, California , USA ; Department of Surgery , University of California , San Diego, California , USA ; Department of Gastroenterological Surgery , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Yong Zhang
- AntiCancer, Inc. , San Diego, California , USA
| | - Shinji Miwa
- AntiCancer, Inc. , San Diego, California , USA ; Department of Surgery , University of California , San Diego, California , USA
| | - Hiroyuki Kishimoto
- Department of Gastroenterological Surgery , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | | | - Michael Bouvet
- Department of Surgery , University of California , San Diego, California , USA
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Robert M Hoffman
- AntiCancer, Inc. , San Diego, California , USA ; Department of Surgery , University of California , San Diego, California , USA
| |
Collapse
|
50
|
Murakami T, Hiroshima Y, Zhang Y, Bouvet M, Chishima T, Tanaka K, Endo I, Hoffman RM. Improved disease-free survival and overall survival after fluorescence-guided surgery of liver metastasis in an orthotopic nude mouse model. J Surg Oncol 2015; 112:119-24. [PMID: 26266663 DOI: 10.1002/jso.23986] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/10/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND In the present study, we sought to determine if fluorescence-guided surgery (FGS) would improve survival compared to standard bright light surgery (BLS) in an experimental colorectal liver metastasis nude mouse model. METHODS Orthotopic nude-mouse models of human HT-29-GFP colon cancer liver metastasis were established in the left lobe of the liver of mice. Fourteen mice with a single liver metastasis were randomized into FGS or BLS groups of seven each. FGS of liver metastasis was performed using a hand-held portable fluorescence imaging system (Dino-Lite) to visualize the GFP fluorescence of the metastasis. The BLS- and FGS-treated mice were followed by weekly fluorescence imaging in order to detect recurrence. RESULTS The bright fluorescence of GFP provided sufficient illumination to accurately distinguish the margins of the metastasis within the liver. Recurrence occurred in multiple sites including the liver, lung, and other organs in the BLS-treated mice but was significantly reduced in FGS-treated mice. The FGS-treated mice had significantly prolonged disease-free survival (P = 0.001) and overall survival (P = 0.027) compared to BLS-treated mice. CONCLUSION The results of the present report demonstrate the feasibility and efficacy of FGS for liver metastasis and suggest its important clinical potential.
Collapse
Affiliation(s)
- Takashi Murakami
- AntiCancer, Inc., San Diego, California.,Department of Surgery, University of California San Diego, San Diego, California.,Department of Gastroenterological Surgery, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Yukihiko Hiroshima
- AntiCancer, Inc., San Diego, California.,Department of Surgery, University of California San Diego, San Diego, California.,Department of Gastroenterological Surgery, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | | | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, California
| | - Takashi Chishima
- Department of Gastroenterological Surgery, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Kuniya Tanaka
- Department of Gastroenterological Surgery, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University, Graduate School of Medicine, Yokohama, Japan
| | - Robert M Hoffman
- AntiCancer, Inc., San Diego, California.,Department of Surgery, University of California San Diego, San Diego, California
| |
Collapse
|